51
|
Vitorakis N, Gargalionis AN, Papavassiliou KA, Adamopoulos C, Papavassiliou AG. Precision Targeting Strategies in Pancreatic Cancer: The Role of Tumor Microenvironment. Cancers (Basel) 2024; 16:2876. [PMID: 39199647 PMCID: PMC11352254 DOI: 10.3390/cancers16162876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 08/14/2024] [Accepted: 08/17/2024] [Indexed: 09/01/2024] Open
Abstract
Pancreatic cancer demonstrates an ever-increasing incidence over the last years and represents one of the top causes of cancer-associated mortality. Cells of the tumor microenvironment (TME) interact with cancer cells in pancreatic ductal adenocarcinoma (PDAC) tumors to preserve cancer cells' metabolism, inhibit drug delivery, enhance immune suppression mechanisms and finally develop resistance to chemotherapy and immunotherapy. New strategies target TME genetic alterations and specific pathways in cell populations of the TME. Complex molecular interactions develop between PDAC cells and TME cell populations including cancer-associated fibroblasts, myeloid-derived suppressor cells, pancreatic stellate cells, tumor-associated macrophages, tumor-associated neutrophils, and regulatory T cells. In the present review, we aim to fully explore the molecular landscape of the pancreatic cancer TME cell populations and discuss current TME targeting strategies to provide thoughts for further research and preclinical testing.
Collapse
Affiliation(s)
- Nikolaos Vitorakis
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Antonios N Gargalionis
- Department of Clinical Biochemistry, 'Attikon' University General Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Kostas A Papavassiliou
- First University Department of Respiratory Medicine, 'Sotiria' Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Christos Adamopoulos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
52
|
Zhang Y, Liu G, Zeng Q, Wu W, Lei K, Zhang C, Tang M, Zhang Y, Xiang X, Tan L, Cui R, Qin S, Song X, Yin C, Chen Z, Kuang M. CCL19-producing fibroblasts promote tertiary lymphoid structure formation enhancing anti-tumor IgG response in colorectal cancer liver metastasis. Cancer Cell 2024; 42:1370-1385.e9. [PMID: 39137726 DOI: 10.1016/j.ccell.2024.07.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 05/05/2024] [Accepted: 07/17/2024] [Indexed: 08/15/2024]
Abstract
Tertiary lymphoid structures (TLSs) are associated with enhanced immunity in tumors. However, their formation and functions in colorectal cancer liver metastasis (CRLM) remain unclear. Here, we reveal that intra- and peri-tumor mature TLSs (TLS+) are associated with improved clinical outcomes than TLS- tumors. Using single-cell-RNA-sequencing and spatial-enhanced-resolution-omics-sequencing (Stereo-seq), we reveal that TLS+ tumors are enriched with IgG+ plasma cells (PCs), while TLS- tumors are characterized with IgA+ PCs. By generating TLS-associated PC-derived monoclonal antibodies in vitro, we show that TLS-PCs secrete tumor-targeting antibodies. As the proof-of-concept, we demonstrate the anti-tumor activities of TLS-PC-mAb6 antibody in humanized mouse model of colorectal cancer. We identify a fibroblast lineage secreting CCL19 that facilitates lymphocyte trafficking to TLSs. CCL19 treatment promotes TLS neogenesis and prevents tumor growth in mice. Our data uncover the central role of CCL19+ fibroblasts in TLS formation, which in turn generates therapeutic antibodies to restrict CRLM.
Collapse
Affiliation(s)
- Yifan Zhang
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Guangjian Liu
- Department of Medical Ultrasonics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Qianwen Zeng
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Wenrui Wu
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Kai Lei
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Chuankai Zhang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Department of Oncology, Cancer Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Miaoling Tang
- Department of Oncology, Cancer Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Yuting Zhang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiao Xiang
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Li Tan
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Rui Cui
- Department of Medical Ultrasonics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Si Qin
- Department of Medical Ultrasonics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Xinming Song
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Changjun Yin
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, 80336 Munich, Germany.
| | - Zhihang Chen
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China.
| | - Ming Kuang
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Sun Yat-sen University Zhongshan School of Medicine, Guangzhou 510080, China.
| |
Collapse
|
53
|
He Z, Li X, Chen S, Cai K, Li X, Liu H. CD105+CAF-derived exosomes CircAMPK1 promotes pancreatic cancer progression by activating autophagy. Exp Hematol Oncol 2024; 13:79. [PMID: 39103892 DOI: 10.1186/s40164-024-00533-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 06/29/2024] [Indexed: 08/07/2024] Open
Abstract
Previous studies have shown that the heterogeneity of tumor-associated fibroblasts (CAFs) in the tumor microenvironment may play a critical role in tumorigenesis; however, the biological function of CAFs in pancreatic cancer is still controversial. In this study, we found that CD105-positive (CD105+) CAF-derived exosomes significantly promoted the proliferative and invasive metastatic abilities of pancreatic cancer cells. Furthermore, RNA-seq and qRT‒PCR experiments revealed circAMPK1 as a key molecule in exosomes from CD105+ CAFs that mediates the malignant progression of pancreatic cancer. Furthermore, we demonstrated that circAMPK1 encodes a novel protein (AMPK1-360aa) in pancreatic cancer cells. This protein competes with AMPK1 to bind to the ubiquitination ligase NEDD4, which inhibits AMPK1 protein degradation and ubiquitination and thereby increases AMPK1 levels. Finally, we demonstrated that AMPK1-360aa induces cellular autophagy via NEDD4/AMPK1 to promote the proliferation and invasion of pancreatic cancer cells. In summary, circAMPK1 in CD105+ CAF-derived exosomes may mediate pancreatic cancer cell proliferation and invasive metastasis by inducing autophagy in target cells. Moreover, circAMPK1 may competitively bind to ubiquitinating enzymes through the encoded protein AMPK1-360aa, which in turn inhibits the ubiquitination-mediated degradation of AMPK1 and contributes to the upregulation of AMPK1 expression, thus inducing cellular autophagy to mediate the malignant progression of pancreatic cancer.
Collapse
Affiliation(s)
- Zhiwei He
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, People's Republic of China
- Department of Hepatobiliary Surgery, Shenzhen University General Hospital & Shenzhen University Clinical Medical Academy Center, Shenzhen University, Shenzhen, 518000, Guangdong, People's Republic of China
| | - Xiushen Li
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, People's Republic of China
| | - Shiyu Chen
- Department of Hepatic-Biliary-Pancreatic Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, People's Republic of China
| | - Kun Cai
- Department of Hepatic-Biliary-Pancreatic Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, People's Republic of China
| | - Xiaowu Li
- Department of Hepatobiliary Surgery, Shenzhen University General Hospital & Shenzhen University Clinical Medical Academy Center, Shenzhen University, Shenzhen, 518000, Guangdong, People's Republic of China.
| | - Hui Liu
- Department of Hepatobiliary Surgery, Shenzhen University General Hospital & Shenzhen University Clinical Medical Academy Center, Shenzhen University, Shenzhen, 518000, Guangdong, People's Republic of China.
| |
Collapse
|
54
|
Inoue C, Miki Y, Saito-Koyama R, Okada Y, Sasano H, Suzuki T. Dipeptidyl peptidase 4-positive cancer-associated fibroblasts enhance lung adenocarcinoma growth. Pathol Res Pract 2024; 260:155418. [PMID: 38908333 DOI: 10.1016/j.prp.2024.155418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/13/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
Cancer-associated fibroblasts (CAFs) are a heterogeneous population of fibroblasts with various features in the cancer stroma and have been reported to influence cancer progression through cell-cell interactions in various types of malignancies, including lung adenocarcinoma (LUAD). Dipeptidyl peptidase 4 (DPP4) is a transmembrane protein with serine protease activity and is involved in the progression of tumors, metabolic diseases, and autoimmune diseases. In the present study, we focused on the role of DPP4-positive CAFs in LUAD. Immunohistochemistry revealed that 38 of 89 LUAD patients showed DPP4 expression in the fibrous stroma, and patients harboring DPP4-positive CAFs were more often male, had a higher Brinkman index, and had a higher Ki-67 labeling index of tumor cells than those with DPP4-negative CAFs. DPP4-positivity was associated with the expression of other CAF markers, α-SMA, periostin, and podoplanin, as well as a cellular senescence marker, p16. In the in vitro study, conditioned media collected from pulmonary fibroblast (OUS-11, HPF, and HPF-C)-induced overexpression of DPP4 significantly promoted the proliferation of LUAD cells (A549 and PC-9) and increased the expression levels of MCP-1, IL-8, IL-6, and GCSF in the media compared to those in controls. In addition, OUS-11 overexpression in DPP4 overexpression increased periostin expression. In conclusion, DPP4-positive CAFs could promote lung adenocarcinoma cell growth by producing soluble factors, and DPP4 inhibition may inhibit cancer progression.
Collapse
Affiliation(s)
- Chihiro Inoue
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Japan.
| | - Yasuhiro Miki
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Japan
| | - Ryoko Saito-Koyama
- Department of Pathology, National Hospital Organization, Sendai Medical Center, Sendai, Japan
| | - Yoshinori Okada
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Hironobu Sasano
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Japan
| | - Takashi Suzuki
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Japan
| |
Collapse
|
55
|
Hagedorn M, Parenti LR, Craddock RA, Comizzoli P, Mabee P, Meinke B, Wolf SM, Bischof JC, Sandlin RD, Tessier SN, Toner M. Safeguarding Earth's biodiversity by creating a lunar biorepository. Bioscience 2024; 74:561-566. [PMID: 39229623 PMCID: PMC11367668 DOI: 10.1093/biosci/biae058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/15/2024] [Accepted: 05/22/2024] [Indexed: 09/05/2024] Open
Abstract
Earth's biodiversity is increasingly threatened and at risk. We propose a passive lunar biorepository for long-term storage of prioritized taxa of live cryopreserved samples to safeguard Earth's biodiversity and to support future space exploration and planet terraforming. Our initial focus will be on cryopreserving animal skin samples with fibroblast cells. An exemplar system has been developed using cryopreserved fish fins from the Starry Goby, Asterropteryx semipunctata. Samples will be expanded into fibroblast cells, recryopreserved, and then tested in an Earth-based laboratory for robust packaging and sensitivity to radiation. Two key factors for this biorepository are the needs to reduce damage from radiation and to maintain the samples near -196° Celsius. Certain lunar sites near the poles may meet these criteria. If possible, further testing would occur on the International Space Station prior to storage on the Moon. To secure a positive shared future, this is an open call to participate in this decades-long program.
Collapse
Affiliation(s)
- Mary Hagedorn
- Smithsonian National Zoo and Conservation Biology Institute, Washington, DC, United States of America
- Hawaii Institute of Marine Biology, Kaneohe, HI, United States of America
| | - Lynne R Parenti
- Smithsonian National Museum of Natural History, Washington, DC, United States of America
| | - Robert A Craddock
- Center for Earth and Planetary Studies, Smithsonian National Air and Space Museum, Washington, DC, United States of America
| | - Pierre Comizzoli
- Smithsonian National Zoo and Conservation Biology Institute, Office of the Smithsonian Under Secretary for Science and Research Washington, DC, United States of America
| | - Paula Mabee
- NEON, Battelle, Boulder, CO, United States of America
| | - Bonnie Meinke
- External Engagement and Business Development, University Corporation for Atmospheric Research (UCAR), Boulder, CO, United States of America
| | - Susan M Wolf
- McKnight Presidential Professor of Law, Medicine & Public Policy; Faegre Drinker Professor of Law, Professor of Medicine at the University of Minnesota, Minneapolis, MN, United States of America
| | - John C Bischof
- McKnight University Professor of Mechanical and Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States of America
| | - Rebecca D Sandlin
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Shannon N Tessier
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Mehmet Toner
- Bioengineering, Massachusetts General Hospital and Harvard Medical School, Shriners Children's Boston, Harvard-MIT Division of Health Sciences and Technology, Boston, MA, United States of America
| |
Collapse
|
56
|
Li E, Cheung HCZ, Ma S. CTHRC1 + fibroblasts and SPP1 + macrophages synergistically contribute to pro-tumorigenic tumor microenvironment in pancreatic ductal adenocarcinoma. Sci Rep 2024; 14:17412. [PMID: 39075108 PMCID: PMC11286765 DOI: 10.1038/s41598-024-68109-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/19/2024] [Indexed: 07/31/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an extremely lethal cancer that accounts for over 90% of all pancreatic cancer cases. With a 5-year survival rate of only 13%, PDAC has proven to be extremely desmoplastic and immunosuppressive to most current therapies, including chemotherapy and surgical resection. In recent years, focus has shifted to understanding the tumor microenvironment (TME) around PDAC, enabling a greater understanding of biological pathways and intercellular interactions that can ultimately lead to potential for future drug targets. In this study, we leverage a combination of single-cell and spatial transcriptomics to further identify cellular populations and interactions within the highly heterogeneous TME. We demonstrate that SPP1+APOE+ tumor-associated macrophages (TAM) and CTHRC1+GREM1+ cancer-associated myofibroblasts (myCAF) not only act synergistically to promote an immune-suppressive TME through active extracellular matrix (ECM) deposition and epithelial mesenchymal transition (EMT), but are spatially colocalized and correlated, leading to worse prognosis. Our results highlight the crosstalk between stromal and myeloid cells as a significant area of study for future therapeutic targets to treat cancer.
Collapse
Affiliation(s)
- Evan Li
- Worcester Academy, Worcester, MA, USA.
| | | | - Shuangge Ma
- Department of Biostatistics, Yale University, New Haven, CT, USA.
| |
Collapse
|
57
|
Wang H, Liang Y, Liu Z, Zhang R, Chao J, Wang M, Liu M, Qiao L, Xuan Z, Zhao H, Lu L. POSTN + cancer-associated fibroblasts determine the efficacy of immunotherapy in hepatocellular carcinoma. J Immunother Cancer 2024; 12:e008721. [PMID: 39067872 PMCID: PMC11284881 DOI: 10.1136/jitc-2023-008721] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2024] [Indexed: 07/30/2024] Open
Abstract
OBJECTIVE Hepatocellular carcinoma (HCC) poses a significant clinical challenge because the long-term benefits of immune checkpoint blockade therapy are limited. A comprehensive understanding of the mechanisms underlying immunotherapy resistance in HCC is imperative for improving patient prognosis. DESIGN In this study, to systematically investigate the characteristics of cancer-associated fibroblast (CAF) subsets and the dynamic communication among the tumor microenvironment (TME) components regulated by CAF subsets, we generated an HCC atlas by compiling single-cell RNA sequencing (scRNA-seq) datasets on 220 samples from six datasets. We combined spatial transcriptomics with scRNA-seq and multiplexed immunofluorescence to identify the specific CAF subsets in the TME that determine the efficacy of immunotherapy in HCC patients. RESULTS Our findings highlight the pivotal role of POSTN+ CAFs as potent immune response barriers at specific tumor locations, as they hinder effective T-cell infiltration and decrease the efficacy of immunotherapy. Additionally, we elucidated the interplay between POSTN+ CAFs and SPP1+ macrophages, whereby the former recruits the latter and triggers increased SPP1 expression via the IL-6/STAT3 signaling pathway. Moreover, we demonstrated a spatial correlation between POSTN+ CAFs and SPP1+ macrophages, revealing an immunosuppressive microenvironment that limits the immunotherapy response. Notably, we found that patients with elevated expression levels of both POSTN+ CAFs and SPP1+ macrophages achieved less therapeutic benefit in an immunotherapy cohort. CONCLUSION Our research elucidates light on the role of a particular subset of CAFs in immunotherapy resistance, emphasizing the potential benefits of targeting specific CAF subpopulations to improve clinical responses to immunotherapy.
Collapse
Affiliation(s)
- Hao Wang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital,Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University & Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China
| | - Yuan Liang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University & Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China
- School of Biological Science & Medical Engineering, Southeast University, Nanjing, Jiangsu, China
| | - Zheng Liu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rui Zhang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University & Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China
| | - Jiashuo Chao
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital,Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University & Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China
| | - Mingming Wang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital,Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University & Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China
| | - Mu Liu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University & Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China
| | - Lei Qiao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University & Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China
| | - Zhengfeng Xuan
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University & Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China
| | - Haitao Zhao
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital,Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Ling Lu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University & Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China
- Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
58
|
Assouline B, Kahn R, Hodali L, Condiotti R, Engel Y, Elyada E, Mordechai-Heyn T, Pitarresi JR, Atias D, Steinberg E, Bidany-Mizrahi T, Forkosh E, Katz LH, Benny O, Golan T, Hofree M, Stewart SA, Atlan KA, Zamir G, Stanger BZ, Berger M, Ben-Porath I. Senescent cancer-associated fibroblasts in pancreatic adenocarcinoma restrict CD8 + T cell activation and limit responsiveness to immunotherapy in mice. Nat Commun 2024; 15:6162. [PMID: 39039076 PMCID: PMC11263607 DOI: 10.1038/s41467-024-50441-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 07/05/2024] [Indexed: 07/24/2024] Open
Abstract
Senescent cells within tumors and their stroma exert complex pro- and anti-tumorigenic functions. However, the identities and traits of these cells, and the potential for improving cancer therapy through their targeting, remain poorly characterized. Here, we identify a senescent subset within previously-defined cancer-associated fibroblasts (CAFs) in pancreatic ductal adenocarcinomas (PDAC) and in premalignant lesions in mice and humans. Senescent CAFs isolated from mouse and humans expressed elevated levels of immune-regulatory genes. Depletion of senescent CAFs, either genetically or using the Bcl-2 inhibitor ABT-199 (venetoclax), increased the proportion of activated CD8+ T cells in mouse pancreatic carcinomas, whereas induction of CAF senescence had the opposite effect. Combining ABT-199 with an immune checkpoint therapy regimen significantly reduced mouse tumor burden. These results indicate that senescent CAFs in PDAC stroma limit the numbers of activated cytotoxic CD8+ T cells, and suggest that their targeted elimination through senolytic treatment may enhance immunotherapy.
Collapse
Grants
- R01 CA217208 NCI NIH HHS
- R01 CA276512 NCI NIH HHS
- R00 CA252153 NCI NIH HHS
- 2621/18 Israel Science Foundation (ISF)
- R01 CA252225 NCI NIH HHS
- R01 AG059244 NIA NIH HHS
- Israel Cancer Research Fund (Israel Cancer Research Fund, Inc.)
- This study was supported by grants from the Israel Science Foundation - Broad Institute program (2621/18, I.B.-P.), Israel Precision Medicine Partnership (3755/21, I.B.-P.), Israel Science Foundation Mid-Career Program (1923/22, I.B.-P.), the Israel Ministry of Science and Technology DKFZ-MOST program (4062, I.B.-P.), the Chief Scientist of the Israel Ministry of Health (3-15017, I.B.-P.), the Alex U. Soyka Program (I.B.-P., B.A., R.K., L.H.), the Israel Cancer Research Fund International Collaboration Program (I.B.-P)
Collapse
Affiliation(s)
- Benjamin Assouline
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rachel Kahn
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Lutfi Hodali
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Reba Condiotti
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yarden Engel
- The Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ela Elyada
- Department of Biochemistry, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Tzlil Mordechai-Heyn
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Surgery, Hadassah Medical Center, and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jason R Pitarresi
- Division of Hematology-Oncology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Molecular Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Dikla Atias
- Pancreatic Cancer Translational Research Laboratory, Oncology Institute, Sheba Medical Center, Tel Hashomer, Israel, and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eliana Steinberg
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Tirza Bidany-Mizrahi
- The Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Esther Forkosh
- Department of Gastroenterology, Hadassah Medical Center, and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Lior H Katz
- Department of Gastroenterology, Hadassah Medical Center, and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ofra Benny
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Talia Golan
- Pancreatic Cancer Translational Research Laboratory, Oncology Institute, Sheba Medical Center, Tel Hashomer, Israel, and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Matan Hofree
- The Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Sheila A Stewart
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Karine A Atlan
- Department of Pathology, Hadassah Medical Center, and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Gideon Zamir
- Department of Surgery, Hadassah Medical Center, and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ben Z Stanger
- Department of Medicine and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Michael Berger
- The Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ittai Ben-Porath
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
59
|
Zheng S, Hu C, Lin Q, Li T, Li G, Tian Q, Zhang X, Huang T, Ye Y, He R, Chen C, Zhou Y, Chen R. Extracellular vesicle-packaged PIAT from cancer-associated fibroblasts drives neural remodeling by mediating m5C modification in pancreatic cancer mouse models. Sci Transl Med 2024; 16:eadi0178. [PMID: 39018369 DOI: 10.1126/scitranslmed.adi0178] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 02/06/2024] [Accepted: 06/25/2024] [Indexed: 07/19/2024]
Abstract
Perineural invasion (PNI) is a biological characteristic commonly observed in pancreatic cancer. Although PNI plays a key role in pancreatic cancer metastasis, recurrence, and poor postoperative survival, its mechanism is largely unclarified. Clinical sample analysis and endoscopic ultrasonographic elasticity scoring indicated that cancer-associated fibroblasts (CAFs) were closely related to the occurrence of PNI. Furthermore, CAF-derived extracellular vesicles (EVs) were involved in PNI in dorsal root ganglion coculture and mouse sciatic nerve models. Next, we demonstrated that CAFs promoted PNI through extracellular vesicle transmission of PNI-associated transcript (PIAT). Mechanistically, PIAT specifically bound to YBX1 and blocked the YBX1-Nedd4l interaction to inhibit YBX1 ubiquitination and degradation. Furthermore, PIAT enhanced the binding of YBX1 and PNI-associated mRNAs in a 5-methylcytosine (m5C)-dependent manner. Mutation of m5C recognition motifs in YBX1 or m5C sites in downstream target genes reversed PIAT-mediated PNI. Consistent with these findings, analyses using a KPC mouse model demonstrated that the PIAT/YBX1 axis enhanced PNI through m5C modification. Clinical data suggested that the PIAT expression in the serum EVs of patients with pancreatic cancer was associated with the degree of neural invasion and prognosis. Our study revealed the important role of the PIAT/YBX1 signaling axis in the tumor microenvironment (TME) in promoting tumor cell PNI and provided a new target for precise interference with CAFs and RNA methylation in the TME to suppress PNI in pancreatic cancer.
Collapse
Affiliation(s)
- Shangyou Zheng
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, People's Republic of China
| | - Chonghui Hu
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, People's Republic of China
| | - Qing Lin
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, People's Republic of China
| | - Tingting Li
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, People's Republic of China
| | - Guolin Li
- Department of Hepatobiliary, Pancreatic and Splenic Surgery, Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510655, People's Republic of China
| | - Qing Tian
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, People's Republic of China
| | - Xiang Zhang
- Department of Gastrointestinal Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, People's Republic of China
| | - Tianhao Huang
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, People's Republic of China
- Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510515, People's Republic of China
| | - Yuancheng Ye
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, People's Republic of China
- Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510515, People's Republic of China
| | - Rihua He
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, People's Republic of China
- Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China
| | - Changhao Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, People's Republic of China
| | - Yu Zhou
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, People's Republic of China
| | - Rufu Chen
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, People's Republic of China
- Department of Hepatobiliary, Pancreatic and Splenic Surgery, Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510655, People's Republic of China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, People's Republic of China
| |
Collapse
|
60
|
Kuhn NF, Zaleta-Linares I, Nyberg WA, Eyquem J, Krummel MF. Localized in vivo gene editing of murine cancer-associated fibroblasts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.11.603114. [PMID: 39071432 PMCID: PMC11275728 DOI: 10.1101/2024.07.11.603114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Discovering the role of fibroblasts residing in the tumor microenvironment (TME) requires controlled, localized perturbations because fibroblasts play critical roles in regulating immunity and tumor biology at multiple sites. Systemic perturbations can lead to unintended, confounding secondary effects, and methods to locally genetically engineer fibroblasts are lacking. To specifically investigate murine stromal cell perturbations restricted to the TME, we developed an adeno-associated virus (AAV)-based method to target any gene-of-interest in fibroblasts at high efficiency (>80%). As proof of concept, we generated single (sKO) and double gene KOs (dKO) of Osmr, Tgfbr2, and Il1r1 in cancer-associated fibroblasts (CAFs) and investigated how their cell states and those of other cells of the TME subsequently change in mouse models of melanoma and pancreatic ductal adenocarcinoma (PDAC). Furthermore, we developed an in vivo knockin-knockout (KIKO) strategy to achieve long-term tracking of CAFs with target gene KO via knocked-in reporter gene expression. This validated in vivo gene editing toolbox is fast, affordable, and modular, and thus holds great potential for further exploration of gene function in stromal cells residing in tumors and beyond.
Collapse
Affiliation(s)
- Nicholas F. Kuhn
- Department of Pathology, University of California, San Francisco, CA 94143, USA
- ImmunoX Initiative, University of California, San Francisco, CA 94143, USA
| | - Itzia Zaleta-Linares
- Department of Pathology, University of California, San Francisco, CA 94143, USA
- ImmunoX Initiative, University of California, San Francisco, CA 94143, USA
| | - William A. Nyberg
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Justin Eyquem
- ImmunoX Initiative, University of California, San Francisco, CA 94143, USA
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Matthew F. Krummel
- Department of Pathology, University of California, San Francisco, CA 94143, USA
- ImmunoX Initiative, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
61
|
Pereira BA, Ritchie S, Chambers CR, Gordon KA, Magenau A, Murphy KJ, Nobis M, Tyma VM, Liew YF, Lucas MC, Naeini MM, Barkauskas DS, Chacon-Fajardo D, Howell AE, Parker AL, Warren SC, Reed DA, Lee V, Metcalf XL, Lee YK, O’Regan LP, Zhu J, Trpceski M, Fontaine ARM, Stoehr J, Rouet R, Lin X, Chitty JL, Porazinski S, Wu SZ, Filipe EC, Cadell AL, Holliday H, Yang J, Papanicolaou M, Lyons RJ, Zaratzian A, Tayao M, Da Silva A, Vennin C, Yin J, Dew AB, McMillan PJ, Goldstein LD, Deveson IW, Croucher DR, Samuel MS, Sim HW, Batten M, Chantrill L, Grimmond SM, Gill AJ, Samra J, Jeffry Evans TR, Sasaki T, Phan TG, Swarbrick A, Sansom OJ, Morton JP, Australian Pancreatic Cancer Matrix Atlas (APMA), Australian Pancreatic Cancer Genome Initiative (APGI), Pajic M, Parker BL, Herrmann D, Cox TR, Timpson P. Temporally resolved proteomics identifies nidogen-2 as a cotarget in pancreatic cancer that modulates fibrosis and therapy response. SCIENCE ADVANCES 2024; 10:eadl1197. [PMID: 38959305 PMCID: PMC11221519 DOI: 10.1126/sciadv.adl1197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 05/30/2024] [Indexed: 07/05/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by increasing fibrosis, which can enhance tumor progression and spread. Here, we undertook an unbiased temporal assessment of the matrisome of the highly metastatic KPC (Pdx1-Cre, LSL-KrasG12D/+, LSL-Trp53R172H/+) and poorly metastatic KPflC (Pdx1-Cre, LSL-KrasG12D/+, Trp53fl/+) genetically engineered mouse models of pancreatic cancer using mass spectrometry proteomics. Our assessment at early-, mid-, and late-stage disease reveals an increased abundance of nidogen-2 (NID2) in the KPC model compared to KPflC, with further validation showing that NID2 is primarily expressed by cancer-associated fibroblasts (CAFs). Using biomechanical assessments, second harmonic generation imaging, and birefringence analysis, we show that NID2 reduction by CRISPR interference (CRISPRi) in CAFs reduces stiffness and matrix remodeling in three-dimensional models, leading to impaired cancer cell invasion. Intravital imaging revealed improved vascular patency in live NID2-depleted tumors, with enhanced response to gemcitabine/Abraxane. In orthotopic models, NID2 CRISPRi tumors had less liver metastasis and increased survival, highlighting NID2 as a potential PDAC cotarget.
Collapse
Affiliation(s)
- Brooke A. Pereira
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
| | - Shona Ritchie
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
| | - Cecilia R. Chambers
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
| | - Katie A. Gordon
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
| | - Astrid Magenau
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
| | - Kendelle J. Murphy
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
| | - Max Nobis
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
- Intravital Imaging Expertise Center, VIB Center for Cancer Biology, VIB, Leuven, Belgium
| | - Victoria M. Tyma
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Ying Fei Liew
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Morghan C. Lucas
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Marjan M. Naeini
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
- Genomics and Inherited Disease Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Deborah S. Barkauskas
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- ACRF INCITe Intravital Imaging Centre, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Diego Chacon-Fajardo
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
- Translational Oncology Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Anna E. Howell
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Amelia L. Parker
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
| | - Sean C. Warren
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
| | - Daniel A. Reed
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
| | - Victoria Lee
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Xanthe L. Metcalf
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Young Kyung Lee
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Luke P. O’Regan
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Jessie Zhu
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
| | - Michael Trpceski
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
| | - Angela R. M. Fontaine
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
- ACRF INCITe Intravital Imaging Centre, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Janett Stoehr
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Romain Rouet
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
- Immune Biotherapies Program, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Xufeng Lin
- Data Science Platform, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Jessica L. Chitty
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
| | - Sean Porazinski
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
- Translational Oncology Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Sunny Z. Wu
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
- Genentech Inc., South San Francisco, CA, USA
| | - Elysse C. Filipe
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
| | - Antonia L. Cadell
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
- Translational Oncology Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Holly Holliday
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington, New South Wales, Australia
| | - Jessica Yang
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
| | - Michael Papanicolaou
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
| | - Ruth J. Lyons
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Anaiis Zaratzian
- Histopathology Platform, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Michael Tayao
- Histopathology Platform, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Andrew Da Silva
- Histopathology Platform, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Claire Vennin
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
- Division of Molecular Pathology, Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands
- Oncode Institute, Amsterdam, Netherlands
| | - Julia Yin
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
- Translational Oncology Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Alysha B. Dew
- Centre for Advanced Histology & Microscopy, Peter MacCallum Cancer Centre, Parkville, Victoria, Australia
| | - Paul J. McMillan
- Centre for Advanced Histology & Microscopy, Peter MacCallum Cancer Centre, Parkville, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- Biological Optical Microscopy Platform, The University of Melbourne, Parkville, Victoria, Australia
| | - Leonard D. Goldstein
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
- Data Science Platform, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Ira W. Deveson
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
- Genomics and Inherited Disease Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - David R. Croucher
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
- Translational Oncology Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Michael S. Samuel
- Centre for Cancer Biology, An Alliance of SA Pathology and University of South Australia, Adelaide, South Australia, Australia
- Basil Hetzel Institute for Translational Health Research, Queen Elizabeth Hospital, Woodville South, South Australia, Australia
| | - Hao-Wen Sim
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, New South Wales, Australia
- Department of Medical Oncology, Chris O’Brien Lifehouse, Camperdown, New South Wales, Australia
| | - Marcel Batten
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Lorraine Chantrill
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- Department of Medical Oncology, Illawarra Shoalhaven Local Health District, Wollongong, New South Wales, Australia
| | - Sean M. Grimmond
- Centre for Cancer Research and Department of Clinical Pathology, The University of Melbourne, Parkville, Victoria, Australia
| | - Anthony J. Gill
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
- Sydney Medical School, University of Sydney, Camperdown, New South Wales, Australia
| | - Jaswinder Samra
- Department of Surgery, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Thomas R. Jeffry Evans
- Cancer Research UK Beatson Institute, Glasgow, UK
- School of Cancer Sciences, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Takako Sasaki
- Department of Biochemistry, Faculty of Medicine, Oita University, Oita, Japan
| | - Tri G. Phan
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
- Precision Immunology Program, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Alexander Swarbrick
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
| | - Owen J. Sansom
- Cancer Research UK Beatson Institute, Glasgow, UK
- School of Cancer Sciences, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Jennifer P. Morton
- Cancer Research UK Beatson Institute, Glasgow, UK
- School of Cancer Sciences, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | | | | | - Marina Pajic
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
- Translational Oncology Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Benjamin L. Parker
- Department of Anatomy and Physiology, University of Melbourne, Parkville, Victoria, Australia
| | - David Herrmann
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
| | - Thomas R. Cox
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
| | - Paul Timpson
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales (UNSW) Sydney, Kensington, New South Wales, Australia
| |
Collapse
|
62
|
Belle JI, Sen D, Baer JM, Liu X, Lander VE, Ye J, Sells BE, Knolhoff BL, Faiz A, Kang LI, Qian G, Fields RC, Ding L, Kim H, Provenzano PP, Stewart SA, DeNardo DG. Senescence Defines a Distinct Subset of Myofibroblasts That Orchestrates Immunosuppression in Pancreatic Cancer. Cancer Discov 2024; 14:1324-1355. [PMID: 38683144 DOI: 10.1158/2159-8290.cd-23-0428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 01/29/2024] [Accepted: 03/08/2024] [Indexed: 05/01/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) therapeutic resistance is largely attributed to a unique tumor microenvironment embedded with an abundance of cancer-associated fibroblasts (CAF). Distinct CAF populations were recently identified, but the phenotypic drivers and specific impact of CAF heterogeneity remain unclear. In this study, we identify a subpopulation of senescent myofibroblastic CAFs (SenCAF) in mouse and human PDAC. These SenCAFs are a phenotypically distinct subset of myofibroblastic CAFs that localize near tumor ducts and accumulate with PDAC progression. To assess the impact of endogenous SenCAFs in PDAC, we used an LSL-KRASG12D;p53flox;p48-CRE;INK-ATTAC (KPPC-IA) mouse model of spontaneous PDAC with inducible senescent cell depletion. Depletion of senescent stromal cells in genetic and pharmacologic PDAC models relieved immune suppression by macrophages, delayed tumor progression, and increased responsiveness to chemotherapy. Collectively, our findings demonstrate that SenCAFs promote PDAC progression and immune cell dysfunction. Significance: CAF heterogeneity in PDAC remains poorly understood. In this study, we identify a novel subpopulation of senescent CAFs that promotes PDAC progression and immunosuppression. Targeting CAF senescence in combination therapies could increase tumor vulnerability to chemo or immunotherapy. See related article by Ye et al., p. 1302.
Collapse
Affiliation(s)
- Jad I Belle
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Devashish Sen
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - John M Baer
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Xiuting Liu
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Varintra E Lander
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Jiayu Ye
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Blake E Sells
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Brett L Knolhoff
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Ahmad Faiz
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Liang-I Kang
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| | - Guhan Qian
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota
- Department of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, Minnesota
| | - Ryan C Fields
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Li Ding
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Hyun Kim
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Paolo P Provenzano
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota
- Department of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, Minnesota
| | - Sheila A Stewart
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| | - David G DeNardo
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
63
|
Zhou Q, Jin X, Zhao Y, Wang Y, Tao M, Cao Y, Yin X. Melanoma-associated fibroblasts in tumor-promotion flammation and antitumor immunity: novel mechanisms and potential immunotherapeutic strategies. Hum Mol Genet 2024; 33:1186-1193. [PMID: 38538564 PMCID: PMC11190611 DOI: 10.1093/hmg/ddae056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 03/09/2024] [Accepted: 03/14/2024] [Indexed: 06/22/2024] Open
Abstract
Melanoma, renowned for its aggressive behavior and resistance to conventional treatments, stands as a formidable challenge in the oncology landscape. The dynamic and complex interplay between cancer cells and the tumor microenvironment has gained significant attention, revealing Melanoma-Associated Fibroblasts (MAFs) as central players in disease progression. The heterogeneity of MAFs endows them with a dual role in melanoma. This exhaustive review seeks to not only shed light on the multifaceted roles of MAFs in orchestrating tumor-promoting inflammation but also to explore their involvement in antitumor immunity. By unraveling novel mechanisms underlying MAF functions, this review aims to provide a comprehensive understanding of their impact on melanoma development. Additionally, it delves into the potential of leveraging MAFs for innovative immunotherapeutic strategies, offering new avenues for enhancing treatment outcomes in the challenging realm of melanoma therapeutics.
Collapse
Affiliation(s)
- Qiujun Zhou
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), #54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang 310000, China
- Department of First Clinical Medical College, Zhejiang Chinese Medical University, #548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310000, China
| | - Xiaoliang Jin
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), #54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang 310000, China
- Department of First Clinical Medical College, Zhejiang Chinese Medical University, #548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310000, China
| | - Ying Zhao
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), #54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang 310000, China
- Department of First Clinical Medical College, Zhejiang Chinese Medical University, #548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310000, China
| | - Yueping Wang
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), #54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang 310000, China
- Department of First Clinical Medical College, Zhejiang Chinese Medical University, #548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310000, China
| | - Maocan Tao
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), #54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang 310000, China
| | - Yi Cao
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), #54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang 310000, China
| | - Xiaohu Yin
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), #54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang 310000, China
| |
Collapse
|
64
|
Xiao Y, Wang Z, Gu M, Wei P, Wang X, Li W. Cancer-associated fibroblasts: heterogeneity and their role in the tumor immune response. Clin Exp Med 2024; 24:126. [PMID: 38864912 PMCID: PMC11169017 DOI: 10.1007/s10238-024-01375-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 05/13/2024] [Indexed: 06/13/2024]
Abstract
In recent decades, many reports have been published on the composition and function of the tumor microenvironment (TME), among which cancer-associated fibroblasts (CAFs) have received much attention. CAFs have different degrees of heterogeneity in terms of their origin, phenotype, and function and can be divided into different subpopulations. These subgroups may play different roles in the occurrence and development of tumors. In addition, CAFs are closely associated with tumor immunity and have been found to regulate immune cell activity and to suppress the tumor immune response. In this review, we systematize the heterogeneity and characteristics of CAFs, discuss how specific CAF subgroups contribute to cancer progression by inducing an immunosuppressive microenvironment, and finally, we examine the future clinical applications of CAF subgroups.
Collapse
Affiliation(s)
- Yuxuan Xiao
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Ziyu Wang
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Meng Gu
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Panjian Wei
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Xiaojue Wang
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Weiying Li
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China.
| |
Collapse
|
65
|
Rhodes JD, Goldenring JR, Lee SH. Regulation of metaplasia and dysplasia in the stomach by the stromal microenvironment. Exp Mol Med 2024; 56:1322-1330. [PMID: 38825636 PMCID: PMC11263556 DOI: 10.1038/s12276-024-01240-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/03/2024] [Accepted: 03/03/2024] [Indexed: 06/04/2024] Open
Abstract
Research on the microenvironment associated with gastric carcinogenesis has focused on cancers of the stomach and often underestimates premalignant stages such as metaplasia and dysplasia. Since epithelial interactions with T cells, macrophages, and type 2 innate lymphoid cells (ILC2s) are indispensable for the formation of precancerous lesions in the stomach, understanding the cellular interactions that promote gastric precancer warrants further investigation. Although various types of immune cells have been shown to play important roles in gastric carcinogenesis, it remains unclear how stromal cells such as fibroblasts influence epithelial transformation in the stomach, especially during precancerous stages. Fibroblasts exist as distinct populations across tissues and perform different functions depending on the expression patterns of cell surface markers and secreted factors. In this review, we provide an overview of known microenvironmental components in the stroma with an emphasis on fibroblast subpopulations and their roles during carcinogenesis in tissues including breast, pancreas, and stomach. Additionally, we offer insights into potential targets of tumor-promoting fibroblasts and identify open areas of research related to fibroblast plasticity and the modulation of gastric carcinogenesis.
Collapse
Affiliation(s)
- Jared D Rhodes
- Program in Cancer Biology, Nashville, TN, USA
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - James R Goldenring
- Program in Cancer Biology, Nashville, TN, USA.
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, USA.
- Section of Surgical Sciences, Nashville, TN, USA.
- Department of Cell and Developmental Biology, Nashville, TN, USA.
- Nashville VA Medical Center, Nashville, TN, USA.
| | - Su-Hyung Lee
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, USA.
- Section of Surgical Sciences, Nashville, TN, USA.
| |
Collapse
|
66
|
Qin Q, Yu R, Eriksson JE, Tsai HI, Zhu H. Cancer-associated fibroblasts in pancreatic ductal adenocarcinoma therapy: Challenges and opportunities. Cancer Lett 2024; 591:216859. [PMID: 38615928 DOI: 10.1016/j.canlet.2024.216859] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 03/30/2024] [Accepted: 04/02/2024] [Indexed: 04/16/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a solid organ malignancy with a high mortality rate. Statistics indicate that its incidence has been increasing as well as the associated deaths. Most patients with PDAC show poor response to therapies making the clinical management of this cancer difficult. Stromal cells in the tumor microenvironment (TME) contribute to the development of resistance to therapy in PDAC cancer cells. Cancer-associated fibroblasts (CAFs), the most prevalent stromal cells in the TME, promote a desmoplastic response, produce extracellular matrix proteins and cytokines, and directly influence the biological behavior of cancer cells. These multifaceted effects make it difficult to eradicate tumor cells from the body. As a result, CAF-targeting synergistic therapeutic strategies have gained increasing attention in recent years. However, due to the substantial heterogeneity in CAF origin, definition, and function, as well as high plasticity, majority of the available CAF-targeting therapeutic approaches are not effective, and in some cases, they exacerbate disease progression. This review primarily elucidates on the effect of CAFs on therapeutic efficiency of various treatment modalities, including chemotherapy, radiotherapy, immunotherapy, and targeted therapy. Strategies for CAF targeting therapies are also discussed.
Collapse
Affiliation(s)
- Qin Qin
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, 212001, China
| | - Rong Yu
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, 212001, China
| | - John E Eriksson
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, FI-20520 Finland
| | - Hsiang-I Tsai
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, 212001, China; Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
| | - Haitao Zhu
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, 212001, China; Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
| |
Collapse
|
67
|
Yamashita K, Kumamoto Y. CAFs-Associated Genes (CAFGs) in Pancreatic Ductal Adenocarcinoma (PDAC) and Novel Therapeutic Strategy. Int J Mol Sci 2024; 25:6003. [PMID: 38892190 PMCID: PMC11172745 DOI: 10.3390/ijms25116003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most aggressive cancer with striking fibrosis, and its mortality rate is ranked second across human cancers. Cancer-associated fibroblasts (CAFs) play a critical role in PDAC progression, and we reviewed the molecular understanding of PDAC CAFs and novel therapeutic potential at present. CAFs-associated genes (CAFGs) were tentatively classified into three categories by stroma specificity representing stroma/epithelia expression ratios (SE ratios). The recent classification using single cell transcriptome technology clarified that CAFs were composed of myofibroblasts (myCAFs), inflammatory CAFs (iCAFs), and other minor ones (e.g., POSTN-CAFs and antigen presenting CAFs, apCAFs). LRRC15 is a myCAFs marker, and myCAFs depletion by diphtheria toxin induces the rapid accumulation of cytotoxic T lymphocytes (CTLs) and therefore augment PDL1 antibody treatments. This finding proposes that myCAFs may be a critical regulator of tumor immunity in terms of PDAC progression. myCAFs are located in CAFs adjacent to tumor cells, while iCAFs marked by PDPN and/or COL14A1 are distant from tumor cells, where hypoxic and acidic environments being located in iCAFs putatively due to poor blood supply is consistent with HIF1A and GPR68 expressions. iCAFs may be shared with SASP (secretion-associated phenotypes) in senescent CAFs. myCAFs are classically characterized by CAFGs induced by TGFB1, while chemoresistant CAFs with SASP may dependent on IL6 expression and accompanied by STAT3 activation. Recently, it was found that the unique metabolism of CAFs can be targeted to prevent PDAC progression, where PDAC cells utilize glucose, whereas CAFs in turn utilize lactate, which may be epigenetically regulated, mediated by its target genes including CXCR4. In summary, CAFs have unique molecular characteristics, which have been rigorously clarified as novel therapeutic targets of PDAC progression.
Collapse
Affiliation(s)
- Keishi Yamashita
- Division of Advanced Surgical Oncology, Research and Development Center for New Medical Frontiers, Kitasato University School of Medicine, Kitasato 1-15-1, Minami-ku, Sagamihara 252-0374, Japan
| | - Yusuke Kumamoto
- Department of General-Pediatric-Hepatobiliary Pancreatic Surgery, Kitasato University School of Medicine, Sagamihara 252-0374, Japan;
| |
Collapse
|
68
|
Liu Y, Han X, Han Y, Bi J, Wu Y, Xiang D, Zhang Y, Bi W, Xu M, Li J. Integrated transcriptomic analysis systematically reveals the heterogeneity and molecular characterization of cancer-associated fibroblasts in osteosarcoma. Gene 2024; 907:148286. [PMID: 38367852 DOI: 10.1016/j.gene.2024.148286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 02/19/2024]
Abstract
BACKGROUND Osteosarcoma (OS), with a peak incidence during the adolescent growth spurt, is correlated with poor prognosis for its high malignancy. The tumor microenvironment (TME) is highly complicated, with frequent interactions between tumor and stromal cells. The cancer-associated fibroblasts (CAFs) in the TME have been considered to actively involve in the progression, metastasis, and drug resistance of OS. This study aimed to characterize cellular heterogeneity and molecular characterization in CAFs subtypes and explore the potential targeting therapeutic strategies to improve the prognosis of OS patients. METHODS The single-cell atlas of human OS tumor lesions were constructed from the GEO database. Then significant marker genes and potential biological functions for each CAFs subtype were identified and explored using the Seurat R package. Next, by performing the survival analyses and constructing the risk scores for CAFs subtypes, we aimed to identify and characterize the prognostic values of specific marker genes and different CAFs subtypes. Furthermore, we explored the therapeutic targets and innovative drugs targeting different CAFs subtypes based on the GDSC database. Finally, prognoses related CAFs subtypes were further validated through immunohistochemistry (IHC) on clinical OS specimens. RESULTS Overall, nine main cell clusters and five subtypes of CAFs were identified. The differentially expressed marker genes for each CAFs clusters were then identified. Moreover, through Gene Ontology (GO) enrichment analysis, we defined the CAFs_2 (upregulated CXCL14 and C3), which was closely related to leukocyte migration and chemotaxis, as inflammatory CAFs (iCAFs). Likewise, we defined the CAFs_4 (upregulated CD74, HLA-DRA and HLA-DRB1), which was closely related to antigen process and presentation, as antigen-presenting CAFs (apCAFs). Furthermore, Kaplan-Meier analyses showed that CAFs_2 and CAFs_4 were correlated with poor clinical prognosis of OS patients. Meanwhile, therapeutic drugs targeting CAFs_2 and CAFs_4, such as 17-AAG/Docetaxel/Bleomycin and PHA-793887/NG-25/KIN001-102, were also explored, respectively. Finally, IHC assay confirmed the abundant CAFs_2 and CAFs_4 subtypes infiltration in the OS microenvironment compared with adjacent tissues. CONCLUSION Our study revealed the diversity, complexity, and heterogeneity of CAFs in OS, and complemented the single-cell atlas in OS TME.
Collapse
Affiliation(s)
- Yuyang Liu
- Department of Neurosurgery, 920th Hospital of Joint Logistics Support Force, Kunming 650032, Yunnan, China; Chinese PLA Spinal Cord Injury Treatment Center, Kunming, Yunnan 650032, China
| | - Xinli Han
- School of Medicine, Nankai University, Tianjin 300074, China
| | - Yuchen Han
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing 100048, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Jingyou Bi
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Yanan Wu
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Dongquan Xiang
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Yinglong Zhang
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Wenzhi Bi
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing 100048, China; School of Medicine, Nankai University, Tianjin 300074, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Meng Xu
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing 100048, China; Medical School of Chinese PLA, Beijing 100853, China.
| | - Jianxiong Li
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing 100048, China.
| |
Collapse
|
69
|
Vendramini-Costa DB, Francescone R, Franco-Barraza J, Luong T, Graves M, de Aquino AM, Steele N, Gardiner JC, Dos Santos SAA, Ogier C, Malloy E, Borghaei L, Martinez E, Zhigarev DI, Tan Y, Lee H, Zhou Y, Cai KQ, Klein-Szanto AJ, Wang H, Andrake M, Dunbrack RL, Campbell K, Cukierman E. Netrin G1 Ligand is a new stromal immunomodulator that promotes pancreatic cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.15.594354. [PMID: 38798370 PMCID: PMC11118300 DOI: 10.1101/2024.05.15.594354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Understanding pancreatic cancer biology is fundamental for identifying new targets and for developing more effective therapies. In particular, the contribution of the stromal microenvironment to pancreatic cancer tumorigenesis requires further exploration. Here, we report the stromal roles of the synaptic protein Netrin G1 Ligand (NGL-1) in pancreatic cancer, uncovering its pro-tumor functions in cancer-associated fibroblasts and in immune cells. We observed that the stromal expression of NGL-1 inversely correlated with patients' overall survival. Moreover, germline knockout (KO) mice for NGL-1 presented decreased tumor burden, with a microenvironment that is less supportive of tumor growth. Of note, tumors from NGL-1 KO mice produced less immunosuppressive cytokines and displayed an increased percentage of CD8 + T cells than those from control mice, while preserving the physical structure of the tumor microenvironment. These effects were shown to be mediated by NGL-1 in both immune cells and in the local stroma, in a TGF-β-dependent manner. While myeloid cells lacking NGL-1 decreased the production of immunosuppressive cytokines, NGL-1 KO T cells showed increased proliferation rates and overall polyfunctionality compared to control T cells. CAFs lacking NGL-1 were less immunosuppressive than controls, with overall decreased production of pro-tumor cytokines and compromised ability to inhibit CD8 + T cells activation. Mechanistically, these CAFs downregulated components of the TGF-β pathway, AP-1 and NFAT transcription factor families, resulting in a less tumor-supportive phenotype. Finally, targeting NGL-1 genetically or using a functionally antagonistic small peptide phenocopied the effects of chemotherapy, while modulating the immunosuppressive tumor microenvironment (TME), rather than eliminating it. We propose NGL-1 as a new local stroma and immunomodulatory molecule, with pro-tumor roles in pancreatic cancer. Statement of Significance Here we uncovered the pro-tumor roles of the synaptic protein NGL-1 in the tumor microenvironment of pancreatic cancer, defining a new target that simultaneously modulates tumor cell, fibroblast, and immune cell functions. This study reports a new pathway where NGL-1 controls TGF-β, AP-1 transcription factor members and NFAT1, modulating the immunosuppressive microenvironment in pancreatic cancer. Our findings highlight NGL-1 as a new stromal immunomodulator in pancreatic cancer.
Collapse
|
70
|
Musiu C, Lupo F, Agostini A, Lionetto G, Bevere M, Paiella S, Carbone C, Corbo V, Ugel S, De Sanctis F. Cellular collusion: cracking the code of immunosuppression and chemo resistance in PDAC. Front Immunol 2024; 15:1341079. [PMID: 38817612 PMCID: PMC11137177 DOI: 10.3389/fimmu.2024.1341079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 05/02/2024] [Indexed: 06/01/2024] Open
Abstract
Despite the efforts, pancreatic ductal adenocarcinoma (PDAC) is still highly lethal. Therapeutic challenges reside in late diagnosis and establishment of peculiar tumor microenvironment (TME) supporting tumor outgrowth. This stromal landscape is highly heterogeneous between patients and even in the same patient. The organization of functional sub-TME with different cellular compositions provides evolutive advantages and sustains therapeutic resistance. Tumor progressively establishes a TME that can suit its own needs, including proliferation, stemness and invasion. Cancer-associated fibroblasts and immune cells, the main non-neoplastic cellular TME components, follow soluble factors-mediated neoplastic instructions and synergize to promote chemoresistance and immune surveillance destruction. Unveiling heterotypic stromal-neoplastic interactions is thus pivotal to breaking this synergism and promoting the reprogramming of the TME toward an anti-tumor milieu, improving thus the efficacy of conventional and immune-based therapies. We underscore recent advances in the characterization of immune and fibroblast stromal components supporting or dampening pancreatic cancer progression, as well as novel multi-omic technologies improving the current knowledge of PDAC biology. Finally, we put into context how the clinic will translate the acquired knowledge to design new-generation clinical trials with the final aim of improving the outcome of PDAC patients.
Collapse
Affiliation(s)
- Chiara Musiu
- Department of Medicine, University of Verona, Verona, Italy
| | - Francesca Lupo
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Antonio Agostini
- Medical Oncology, Department of Translational Medicine, Catholic University of the Sacred Heart, Rome, Italy
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Gabriella Lionetto
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Michele Bevere
- ARC-Net Research Centre, University of Verona, Verona, Italy
| | - Salvatore Paiella
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Carmine Carbone
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Vincenzo Corbo
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Stefano Ugel
- Department of Medicine, University of Verona, Verona, Italy
| | | |
Collapse
|
71
|
Niu N, Shen X, Wang Z, Chen Y, Weng Y, Yu F, Tang Y, Lu P, Liu M, Wang L, Sun Y, Yang M, Shen B, Jin J, Lu Z, Jiang K, Shi Y, Xue J. Tumor cell-intrinsic epigenetic dysregulation shapes cancer-associated fibroblasts heterogeneity to metabolically support pancreatic cancer. Cancer Cell 2024; 42:869-884.e9. [PMID: 38579725 DOI: 10.1016/j.ccell.2024.03.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 02/01/2024] [Accepted: 03/11/2024] [Indexed: 04/07/2024]
Abstract
The tumor microenvironment (TME) in pancreatic ductal adenocarcinoma (PDAC) involves a significant accumulation of cancer-associated fibroblasts (CAFs) as part of the host response to tumor cells. The origins and functions of transcriptionally diverse CAF populations in PDAC remain poorly understood. Tumor cell-intrinsic genetic mutations and epigenetic dysregulation may reshape the TME; however, their impacts on CAF heterogeneity remain elusive. SETD2, a histone H3K36 trimethyl-transferase, functions as a tumor suppressor. Through single-cell RNA sequencing, we identify a lipid-laden CAF subpopulation marked by ABCA8a in Setd2-deficient pancreatic tumors. Our findings reveal that tumor-intrinsic SETD2 loss unleashes BMP2 signaling via ectopic gain of H3K27Ac, leading to CAFs differentiation toward lipid-rich phenotype. Lipid-laden CAFs then enhance tumor progression by providing lipids for mitochondrial oxidative phosphorylation via ABCA8a transporter. Together, our study links CAF heterogeneity to epigenetic dysregulation in tumor cells, highlighting a previously unappreciated metabolic interaction between CAFs and pancreatic tumor cells.
Collapse
Affiliation(s)
- Ningning Niu
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuqing Shen
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yueyue Chen
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yawen Weng
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feier Yu
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingying Tang
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping Lu
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingzhu Liu
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liwei Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yongwei Sun
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Minwei Yang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baiyong Shen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiabin Jin
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zipeng Lu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Kuirong Jiang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Yufeng Shi
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, China
| | - Jing Xue
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
72
|
Jalil SMA, Henry JC, Cameron AJM. Targets in the Tumour Matrisome to Promote Cancer Therapy Response. Cancers (Basel) 2024; 16:1847. [PMID: 38791926 PMCID: PMC11119821 DOI: 10.3390/cancers16101847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
The extracellular matrix (ECM) is composed of complex fibrillar proteins, proteoglycans, and macromolecules, generated by stromal, immune, and cancer cells. The components and organisation of the matrix evolves as tumours progress to invasive disease and metastasis. In many solid tumours, dense fibrotic ECM has been hypothesised to impede therapy response by limiting drug and immune cell access. Interventions to target individual components of the ECM, collectively termed the matrisome, have, however, revealed complex tumour-suppressor, tumour-promoter, and immune-modulatory functions, which have complicated clinical translation. The degree to which distinct components of the matrisome can dictate tumour phenotypes and response to therapy is the subject of intense study. A primary aim is to identify therapeutic opportunities within the matrisome, which might support a better response to existing therapies. Many matrix signatures have been developed which can predict prognosis, immune cell content, and immunotherapy responses. In this review, we will examine key components of the matrisome which have been associated with advanced tumours and therapy resistance. We have primarily focussed here on targeting matrisome components, rather than specific cell types, although several examples are described where cells of origin can dramatically affect tumour roles for matrix components. As we unravel the complex biochemical, biophysical, and intracellular transduction mechanisms associated with the ECM, numerous therapeutic opportunities will be identified to modify tumour progression and therapy response.
Collapse
Affiliation(s)
| | | | - Angus J. M. Cameron
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK; (S.M.A.J.); (J.C.H.)
| |
Collapse
|
73
|
Lin S, Zhou M, Cheng L, Shuai Z, Zhao M, Jie R, Wan Q, Peng F, Ding S. Exploring the association of POSTN + cancer-associated fibroblasts with triple-negative breast cancer. Int J Biol Macromol 2024; 268:131560. [PMID: 38631570 DOI: 10.1016/j.ijbiomac.2024.131560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024]
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer with a poor prognosis. Cancer-associated fibroblasts (CAFs) play a critical role in regulating TNBC tumor development. This study aimed to identify and characterize a specific subtype of CAFs associated with TNBC. Initially, using high-throughput bulk transcriptomic data in two cohorts, we identified three CAF-related subtypes (CS1, CS2, CS3) in TNBC samples. These three CAFs subtypes were closely linked to the tumor microenvironment. The CS1 subtype exhibited a relatively immune-rich microenvironment and a favourable prognosis, whereas the CS3 subtype displayed an immune-deprived tumor microenvironment and an unfavourable prognosis. Through WGCNA analysis, POSTN was identified as a key biomarker for CAFs associated with TNBC. Then, POSTN+CAFs was identified and characterized. Both POSTN and POSTN+CAFs showed significant positive correlations with stromal molecules HGF and MET at both the transcriptional and protein levels. Specifically co-localized with CAFs in the tumor stromal area, POSTN, produced by POSTN+CAFs, could modulate the HGF-MET axis, serving as a bypass activation pathway to regulate tumor cell proliferation in response to EGFR inhibitor and MET inhibitor. This study underscores the significance of POSTN and POSTN+CAFs as crucial targets for the diagnosis and treatment of TNBC.
Collapse
Affiliation(s)
- Shuangyan Lin
- Department of Cell Biology and Department of Cardiovascular Surgery, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 57 Zhugan Lane, Hangzhou 310000, Zhejiang, China; Department of Pathology, Zhejiang Hospital, Zhejiang University School of Medicine, 12 Lingyin Rd, Hangzhou 310013, Zhejiang, China
| | - Miaoni Zhou
- Department of Dermatology, Hangzhou Third People's Hospital, Affiliated Hangzhou Dermatology Hospital, Zhejiang University School of Medicine, 38 Xihu Rd, Hangzhou 310009, Zhejiang, China
| | - Liying Cheng
- Jiaxing University Medical College, 899 Shiguang Rd, Jiaxing 314001, Zhejiang, China
| | - Zhifeng Shuai
- Department of Pathology, Zhejiang Hospital, 12 Lingyin Rd, Hangzhou 310013, Zhejiang, China
| | - Mingyuan Zhao
- Department of Pathology, Zhejiang Hospital, 12 Lingyin Rd, Hangzhou 310013, Zhejiang, China
| | - Ruixia Jie
- Department of Pathology, Zhejiang Hospital, 12 Lingyin Rd, Hangzhou 310013, Zhejiang, China
| | - Qun Wan
- Department of Urinary Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, Zhejiang, China
| | - Fang Peng
- Department of Pathology, Zhejiang Hospital, 12 Lingyin Rd, Hangzhou 310013, Zhejiang, China.
| | - Shiping Ding
- Department of Cell Biology and Department of Cardiovascular Surgery, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 57 Zhugan Lane, Hangzhou 310000, Zhejiang, China; Department of Cell Biology, Zhejiang University School of Medicine, 866 Yuhangtang Rd, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
74
|
Sun W, Xie S, Liu SF, Hu X, Xing D. Evolving Tumor Characteristics and Smart Nanodrugs for Tumor Immunotherapy. Int J Nanomedicine 2024; 19:3919-3942. [PMID: 38708176 PMCID: PMC11070166 DOI: 10.2147/ijn.s453265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/11/2024] [Indexed: 05/07/2024] Open
Abstract
Typical physiological characteristics of tumors, such as weak acidity, low oxygen content, and upregulation of certain enzymes in the tumor microenvironment (TME), provide survival advantages when exposed to targeted attacks by drugs and responsive nanomedicines. Consequently, cancer treatment has significantly progressed in recent years. However, the evolution and adaptation of tumor characteristics still pose many challenges for current treatment methods. Therefore, efficient and precise cancer treatments require an understanding of the heterogeneity degree of various factors in cancer cells during tumor evolution to exploit the typical TME characteristics and manage the mutation process. The highly heterogeneous tumor and infiltrating stromal cells, immune cells, and extracellular components collectively form a unique TME, which plays a crucial role in tumor malignancy, including proliferation, invasion, metastasis, and immune escape. Therefore, the development of new treatment methods that can adapt to the evolutionary characteristics of tumors has become an intense focus in current cancer treatment research. This paper explores the latest understanding of cancer evolution, focusing on how tumors use new antigens to shape their "new faces"; how immune system cells, such as cytotoxic T cells, regulatory T cells, macrophages, and natural killer cells, help tumors become "invisible", that is, immune escape; whether the diverse cancer-associated fibroblasts provide support and coordination for tumors; and whether it is possible to attack tumors in reverse. This paper discusses the limitations of targeted therapy driven by tumor evolution factors and explores future strategies and the potential of intelligent nanomedicines, including the systematic coordination of tumor evolution factors and adaptive methods, to meet this therapeutic challenge.
Collapse
Affiliation(s)
- Wenshe Sun
- The Affiliated Hospital of Qingdao University, Qingdao, 266071, People’s Republic of China
- Qingdao Cancer Institute, Qingdao University, Qingdao, 266071, People’s Republic of China
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, People’s Republic of China
| | - Shaowei Xie
- Department of Ultrasound, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People’s Republic of China
| | - Shi Feng Liu
- The Affiliated Hospital of Qingdao University, Qingdao, 266071, People’s Republic of China
| | - Xiaokun Hu
- The Affiliated Hospital of Qingdao University, Qingdao, 266071, People’s Republic of China
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao, 266071, People’s Republic of China
- Qingdao Cancer Institute, Qingdao University, Qingdao, 266071, People’s Republic of China
| |
Collapse
|
75
|
Raymant M, Astuti Y, Alvaro-Espinosa L, Green D, Quaranta V, Bellomo G, Glenn M, Chandran-Gorner V, Palmer DH, Halloran C, Ghaneh P, Henderson NC, Morton JP, Valiente M, Mielgo A, Schmid MC. Macrophage-fibroblast JAK/STAT dependent crosstalk promotes liver metastatic outgrowth in pancreatic cancer. Nat Commun 2024; 15:3593. [PMID: 38678021 PMCID: PMC11055860 DOI: 10.1038/s41467-024-47949-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 04/16/2024] [Indexed: 04/29/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly metastatic disease for which better therapies are urgently needed. Fibroblasts and macrophages are heterogeneous cell populations able to enhance metastasis, but the role of a macrophage-fibroblast crosstalk in regulating their pro-metastatic functions remains poorly understood. Here we deconvolve how macrophages regulate metastasis-associated fibroblast (MAF) heterogeneity in the liver. We identify three functionally distinct MAF populations, among which the generation of pro-metastatic and immunoregulatory myofibroblastic-MAFs (myMAFs) critically depends on macrophages. Mechanistically, myMAFs are induced through a STAT3-dependent mechanism driven by macrophage-derived progranulin and cancer cell-secreted leukaemia inhibitory factor (LIF). In a reciprocal manner, myMAF secreted osteopontin promotes an immunosuppressive macrophage phenotype resulting in the inhibition of cytotoxic T cell functions. Pharmacological blockade of STAT3 or myMAF-specific genetic depletion of STAT3 restores an anti-tumour immune response and reduces metastases. Our findings provide molecular insights into the complex macrophage-fibroblast interactions in tumours and reveal potential targets to inhibit PDAC liver metastasis.
Collapse
Affiliation(s)
- Meirion Raymant
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Yuliana Astuti
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Laura Alvaro-Espinosa
- Brain Metastasis Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Daniel Green
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Valeria Quaranta
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Gaia Bellomo
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Mark Glenn
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Vatshala Chandran-Gorner
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Daniel H Palmer
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Christopher Halloran
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Paula Ghaneh
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Neil C Henderson
- Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, EH16 4TJ, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Jennifer P Morton
- Cancer Research-UK Scotland Institute and School of Cancer Sciences, University of Glasgow, Switchback Road, Glasgow, G61 1BD, UK
| | - Manuel Valiente
- Brain Metastasis Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Ainhoa Mielgo
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Michael C Schmid
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK.
| |
Collapse
|
76
|
Zabransky DJ, Chhabra Y, Fane ME, Kartalia E, Leatherman JM, Hüser L, Zimmerman JW, Delitto D, Han S, Armstrong TD, Charmsaz S, Guinn S, Pramod S, Thompson ED, Hughes SJ, O’Connell J, Egan JM, Jaffee EM, Weeraratna AT. Fibroblasts in the Aged Pancreas Drive Pancreatic Cancer Progression. Cancer Res 2024; 84:1221-1236. [PMID: 38330147 PMCID: PMC11835372 DOI: 10.1158/0008-5472.can-24-0086] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/10/2024]
Abstract
UNLABELLED Pancreatic cancer is more prevalent in older individuals and often carries a poorer prognosis for them. The relationship between the microenvironment and pancreatic cancer is multifactorial, and age-related changes in nonmalignant cells in the tumor microenvironment may play a key role in promoting cancer aggressiveness. Because fibroblasts have profound impacts on pancreatic cancer progression, we investigated whether age-related changes in pancreatic fibroblasts influence cancer growth and metastasis. Proteomics analysis revealed that aged fibroblasts secrete different factors than young fibroblasts, including increased growth/differentiation factor 15 (GDF-15). Treating young mice with GDF-15 enhanced tumor growth, whereas aged GDF-15 knockout mice showed reduced tumor growth. GDF-15 activated AKT, rendering tumors sensitive to AKT inhibition in an aged but not young microenvironment. These data provide evidence for how aging alters pancreatic fibroblasts and promotes tumor progression, providing potential therapeutic targets and avenues for studying pancreatic cancer while accounting for the effects of aging. SIGNIFICANCE Aged pancreatic fibroblasts secrete GDF-15 and activate AKT signaling to promote pancreatic cancer growth, highlighting the critical role of aging-mediated changes in the pancreatic cancer microenvironment in driving tumor progression. See related commentary by Isaacson et al., p. 1185.
Collapse
Affiliation(s)
- Daniel J. Zabransky
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Yash Chhabra
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205
| | - Mitchell E. Fane
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205
- Fox Chase Cancer Center, Cancer Signaling and Microenvironment Program, Philadelphia, PA 19001
| | - Emma Kartalia
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - James M. Leatherman
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Laura Hüser
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Jacquelyn W. Zimmerman
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Daniel Delitto
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305; Department of Surgery, Stanford University School of Medicine, Stanford CA 94305
| | - Song Han
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610
| | - Todd D. Armstrong
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Soren Charmsaz
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Samantha Guinn
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Sneha Pramod
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205
| | - Elizabeth D. Thompson
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Steven J. Hughes
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610
| | - Jennifer O’Connell
- Diabetes Section/Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224
| | - Josephine M. Egan
- Diabetes Section/Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224
| | - Elizabeth M. Jaffee
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- The Johns Hopkins Cancer Convergence Institute, Baltimore, MD, 21287
| | - Ashani T. Weeraratna
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205
| |
Collapse
|
77
|
Boelaars K, Rodriguez E, Huinen ZR, Liu C, Wang D, Springer BO, Olesek K, Goossens-Kruijssen L, van Ee T, Lindijer D, Tak W, de Haas A, Wehry L, Nugteren-Boogaard JP, Mikula A, de Winde CM, Mebius RE, Tuveson DA, Giovannetti E, Bijlsma MF, Wuhrer M, van Vliet SJ, van Kooyk Y. Pancreatic cancer-associated fibroblasts modulate macrophage differentiation via sialic acid-Siglec interactions. Commun Biol 2024; 7:430. [PMID: 38594506 PMCID: PMC11003967 DOI: 10.1038/s42003-024-06087-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/21/2024] [Indexed: 04/11/2024] Open
Abstract
Despite recent advances in cancer immunotherapy, pancreatic ductal adenocarcinoma (PDAC) remains unresponsive due to an immunosuppressive tumor microenvironment, which is characterized by the abundance of cancer-associated fibroblasts (CAFs). Once identified, CAF-mediated immune inhibitory mechanisms could be exploited for cancer immunotherapy. Siglec receptors are increasingly recognized as immune checkpoints, and their ligands, sialic acids, are known to be overexpressed by cancer cells. Here, we unveil a previously unrecognized role of sialic acid-containing glycans on PDAC CAFs as crucial modulators of myeloid cells. Using multiplex immunohistochemistry and transcriptomics, we show that PDAC stroma is enriched in sialic acid-containing glycans compared to tumor cells and normal fibroblasts, and characterized by ST3GAL4 expression. We demonstrate that sialic acids on CAF cell lines serve as ligands for Siglec-7, -9, -10 and -15, distinct from the ligands on tumor cells, and that these receptors are found on myeloid cells in the stroma of PDAC biopsies. Furthermore, we show that CAFs drive the differentiation of monocytes to immunosuppressive tumor-associated macrophages in vitro, and that CAF sialylation plays a dominant role in this process compared to tumor cell sialylation. Collectively, our findings unravel sialic acids as a mechanism of CAF-mediated immunomodulation, which may provide targets for immunotherapy in PDAC.
Collapse
Affiliation(s)
- Kelly Boelaars
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, De Boelelaan, 1117, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - Ernesto Rodriguez
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, De Boelelaan, 1117, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - Zowi R Huinen
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, De Boelelaan, 1117, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - Chang Liu
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, De Boelelaan, 1117, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
- Amsterdam UMC location Vrije Universiteit Amsterdam, Pulmonary Medicine, De Boelelaan, 1117, Amsterdam, the Netherlands
| | - Di Wang
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Babet O Springer
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, De Boelelaan, 1117, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - Katarzyna Olesek
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, De Boelelaan, 1117, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - Laura Goossens-Kruijssen
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, De Boelelaan, 1117, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - Thomas van Ee
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, De Boelelaan, 1117, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - Dimitri Lindijer
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, De Boelelaan, 1117, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - Willemijn Tak
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, De Boelelaan, 1117, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - Aram de Haas
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, De Boelelaan, 1117, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - Laetitia Wehry
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, De Boelelaan, 1117, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - Joline P Nugteren-Boogaard
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, De Boelelaan, 1117, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - Aleksandra Mikula
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, De Boelelaan, 1117, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - Charlotte M de Winde
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, De Boelelaan, 1117, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - Reina E Mebius
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, De Boelelaan, 1117, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | | | - Elisa Giovannetti
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam UMC location Vrije Universiteit Amsterdam, Medical Oncology, De Boelelaan, 1117, Amsterdam, Netherlands
- Cancer Pharmacology Lab, AIRC Start-Up Unit, Fondazione Pisana per la Scienza, Pisa, Italy
| | - Maarten F Bijlsma
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam UMC, location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory for Experimental Oncology and Radiobiology, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Manfred Wuhrer
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Sandra J van Vliet
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, De Boelelaan, 1117, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - Yvette van Kooyk
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, De Boelelaan, 1117, Amsterdam, Netherlands.
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands.
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands.
| |
Collapse
|
78
|
Gao Z, Zhang N, An B, Li D, Fang Z, Xu D. Comprehensive analyses of the cancer-associated fibroblast subtypes and their score system for prediction of outcomes and immunosuppressive microenvironment in prostate cancer. Cancer Cell Int 2024; 24:127. [PMID: 38580966 PMCID: PMC10996219 DOI: 10.1186/s12935-024-03305-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 03/19/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) drive cancer progression and treatment failure on one hand, while their tumor-restraining functions are also observed on the other. Recent single cell RNA sequencing (scRNA-seq) analyses demonstrates heterogeneity of CAFs and defines molecular subtypes of CAFs, which help explain their different functions. However, it remains unclear whether these CAF subtypes have the same or different biological/clinical implications in prostate cancer (PCa) or other malignancies. METHODS PCa cells were incubated with supernatant from normal fibroblasts and CAFs to assess their effects on cell behaviors. Sequencing, genomic, and clinical data were collected from TCGA, MSKCC, CPGEA and GEO databases. CAF molecular subtypes and total CAF scores were constructed and grouped into low and high groups based on CAF-specific gene expression. Progression free interval (PFI), clinicopathological features, telomere length, immune cell infiltration, drug treatment and somatic mutations were compared among CAF molecular subtypes and low/high score groups. RESULTS The PCa CAF-derived supernatant promoted PCa cell proliferation and invasion. Based on differentially expressed genes identified by scRNA-seq analyses, we classified CAFs into 6 molecular subtypes in PCa tumors, and each subtype was then categorized into score-high and low groups according to the subtype-specific gene expression level. Such score models in 6 CAF subtypes all predicted PFI. Telomeres were significantly shorter in high-score tumors. The total CAF score from 6 CAF subtypes was also associated with PFI in PCa patients inversely, which was consistent with results from cellular experiments. Immunosuppressive microenvironment occurred more frequently in tumors with a high CAF score, which was characterized by increased CTLA4 expression and indicated better responses to CTLA4 inhibitors. Moreover, this model can also serve as a useful PFI predictor in pan-cancers. CONCLUSION By combining scRNA-seq and bulk RNA-seq data analyses, we develop a CAF subtype score system as a prognostic factor for PCa and other cancer types. This model system also helps distinguish different immune-suppressive mechanisms in PCa, suggesting its implications in predicting response to immunotherapy. Thus, the present findings should contribute to personalized PCa intervention.
Collapse
Affiliation(s)
- Ze Gao
- Department of Urology, Qilu Hospital of Shandong University, Jinan, 250012, China
- Institute of Andrology, Shandong University, Jinan, 250012, China
| | - Ning Zhang
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Bingzheng An
- Department of Urology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Dawei Li
- Department of Urology, Qilu Hospital of Shandong University, Jinan, 250012, China
- Institute of Andrology, Shandong University, Jinan, 250012, China
| | - Zhiqing Fang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, 250012, China.
- Institute of Andrology, Shandong University, Jinan, 250012, China.
| | - Dawei Xu
- Department of Medicine, Division of Hematology, Bioclinicum, Karolinska Institute and, Karolinska University Hospital, Solna, Stockholm, SE-17176, Sweden.
| |
Collapse
|
79
|
Lloyd EG, Henríquez JA, Biffi G. Modelling the micro- and macro- environment of pancreatic cancer: from patients to pre-clinical models and back. Dis Model Mech 2024; 17:dmm050624. [PMID: 38639944 PMCID: PMC11051978 DOI: 10.1242/dmm.050624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy with very low survival rates. Over the past 50 years, improvements in PDAC survival have significantly lagged behind the progress made in other cancers. PDAC's dismal prognosis is due to typical late-stage diagnosis combined with lack of effective treatments and complex mechanisms of disease. We propose that improvements in survival are partly hindered by the current focus on largely modelling and targeting PDAC as one disease, despite it being heterogeneous. Implementing new disease-representative pre-clinical mouse models that capture this complexity could enable the development of transformative therapies. Specifically, these models should recapitulate human PDAC late-stage biology, heterogeneous genetics, extensive non-malignant stroma, and associated risk factors and comorbidities. In this Perspective, we focus on how pre-clinical mouse models could be improved to exemplify key features of PDAC micro- and macro- environments, which would drive clinically relevant patient stratification, tailored treatments and improved survival.
Collapse
Affiliation(s)
- Eloise G. Lloyd
- University of Cambridge, Cancer Research UK Cambridge Institute, Robinson Way, Cambridge CB2 0RE, UK
| | - Joaquín Araos Henríquez
- University of Cambridge, Cancer Research UK Cambridge Institute, Robinson Way, Cambridge CB2 0RE, UK
| | - Giulia Biffi
- University of Cambridge, Cancer Research UK Cambridge Institute, Robinson Way, Cambridge CB2 0RE, UK
| |
Collapse
|
80
|
Huang H, Lu W, Zhang X, Pan J, Cao F, Wen L. Fibroblast subtypes in pancreatic cancer and pancreatitis: from mechanisms to therapeutic strategies. Cell Oncol (Dordr) 2024; 47:383-396. [PMID: 37721678 DOI: 10.1007/s13402-023-00874-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2023] [Indexed: 09/19/2023] Open
Abstract
Excessive fibrosis is a predominant feature of pancreatic stroma and plays a crucial role in the development and progression of pancreatic ductal adenocarcinoma (PDAC) and chronic pancreatitis (CP). Emerging evidence showed diversity and heterogeneity of fibroblasts play crucial and somewhat contradictory roles, the interactions between fibroblasts and pancreatic cells or infiltrating immune cells are of great importance during PDAC and CP progression, with some promising therapeutic strategies being tested. Therefore, in this review, we describe the classification of fibroblasts and their functions in PDAC and pancreatitis, the mechanisms by which fibroblasts mediate the development and progression of PDAC and CP through direct or indirect interaction between fibroblast and pancreatic parenchymal cells, or by remodeling the pancreatic immune microenvironment mediates the development and progression of PDAC and CP. Finally, we summarized the current therapeutic strategies and agents that directly target subtypes of fibroblasts or interfere with their essential functions.
Collapse
Affiliation(s)
- Huizhen Huang
- Department of Gastroenterology, Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Nanjing Medical University, Shanghai, China
| | - Wanyi Lu
- Center for Biomarker Discovery and Validation, National Infrastructures for Translational Medicine (PUMCH), Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Xiuli Zhang
- Center for Biomarker Discovery and Validation, National Infrastructures for Translational Medicine (PUMCH), Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Jiachun Pan
- Center for Biomarker Discovery and Validation, National Infrastructures for Translational Medicine (PUMCH), Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Feng Cao
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Li Wen
- Center for Biomarker Discovery and Validation, National Infrastructures for Translational Medicine (PUMCH), Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China.
- State Key Laboratory of Complex, Severe, and Rare Diseases, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China.
| |
Collapse
|
81
|
Zhu J, Yang W, Ma J, He H, Liu Z, Zhu X, He X, He J, Chen Z, Jin X, Wang X, He K, Wei W, Hu J. Pericyte signaling via soluble guanylate cyclase shapes the vascular niche and microenvironment of tumors. EMBO J 2024; 43:1519-1544. [PMID: 38528180 PMCID: PMC11021551 DOI: 10.1038/s44318-024-00078-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/04/2024] [Accepted: 03/07/2024] [Indexed: 03/27/2024] Open
Abstract
Pericytes and endothelial cells (ECs) constitute the fundamental components of blood vessels. While the role of ECs in tumor angiogenesis and the tumor microenvironment is well appreciated, pericyte function in tumors remains underexplored. In this study, we used pericyte-specific deletion of the nitric oxide (NO) receptor, soluble guanylate cyclase (sGC), to investigate via single-cell RNA sequencing how pericytes influence the vascular niche and the tumor microenvironment. Our findings demonstrate that pericyte sGC deletion disrupts EC-pericyte interactions, impairing Notch-mediated intercellular communication and triggering extensive transcriptomic reprogramming in both pericytes and ECs. These changes further extended their influence to neighboring cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs) through paracrine signaling, collectively suppressing tumor growth. Inhibition of pericyte sGC has minimal impact on quiescent vessels but significantly increases the vulnerability of angiogenic tumor vessels to conventional anti-angiogenic therapy. In conclusion, our findings elucidate the role of pericytes in shaping the tumor vascular niche and tumor microenvironment and support pericyte sGC targeting as a promising strategy for improving anti-angiogenic therapy for cancer treatment.
Collapse
Affiliation(s)
- Jing Zhu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Wu Yang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Jianyun Ma
- University of Chinese Academy of Sciences, Beijing, China
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Hao He
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhen Liu
- University of Chinese Academy of Sciences, Beijing, China
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
- Lingang Laboratory, Shanghai, China
| | - Xiaolan Zhu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xueyang He
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jing He
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhan Chen
- Pathology Department, Cixi People's Hospital, Zhejiang, China
| | - Xiaoliang Jin
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaohong Wang
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Kaiwen He
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Shanghai Key Laboratory of Aging Studies, Shanghai, China
| | - Wu Wei
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China.
- Lingang Laboratory, Shanghai, China.
| | - Junhao Hu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Shanghai Key Laboratory of Aging Studies, Shanghai, China.
| |
Collapse
|
82
|
Broz MT, Ko EY, Ishaya K, Xiao J, De Simone M, Hoi XP, Piras R, Gala B, Tessaro FHG, Karlstaedt A, Orsulic S, Lund AW, Chan KS, Guarnerio J. Metabolic targeting of cancer associated fibroblasts overcomes T-cell exclusion and chemoresistance in soft-tissue sarcomas. Nat Commun 2024; 15:2498. [PMID: 38509063 PMCID: PMC10954767 DOI: 10.1038/s41467-024-46504-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 02/29/2024] [Indexed: 03/22/2024] Open
Abstract
T cell-based immunotherapies have exhibited promising outcomes in tumor control; however, their efficacy is limited in immune-excluded tumors. Cancer-associated fibroblasts (CAFs) play a pivotal role in shaping the tumor microenvironment and modulating immune infiltration. Despite the identification of distinct CAF subtypes using single-cell RNA-sequencing (scRNA-seq), their functional impact on hindering T-cell infiltration remains unclear, particularly in soft-tissue sarcomas (STS) characterized by low response rates to T cell-based therapies. In this study, we characterize the STS microenvironment using murine models (in female mice) with distinct immune composition by scRNA-seq, and identify a subset of CAFs we termed glycolytic cancer-associated fibroblasts (glyCAF). GlyCAF rely on GLUT1-dependent expression of CXCL16 to impede cytotoxic T-cell infiltration into the tumor parenchyma. Targeting glycolysis decreases T-cell restrictive glyCAF accumulation at the tumor margin, thereby enhancing T-cell infiltration and augmenting the efficacy of chemotherapy. These findings highlight avenues for combinatorial therapeutic interventions in sarcomas and possibly other solid tumors. Further investigations and clinical trials are needed to validate these potential strategies and translate them into clinical practice.
Collapse
Affiliation(s)
- Marina T Broz
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Emily Y Ko
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kristin Ishaya
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jinfen Xiao
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Marco De Simone
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Xen Ping Hoi
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Roberta Piras
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Basia Gala
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Fernando H G Tessaro
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Anja Karlstaedt
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- David Geffen Medical School, Department of Medicine, University of California, Los Angeles, CA, USA
| | - Sandra Orsulic
- David Geffen Medical School, Department of Medicine, University of California, Los Angeles, CA, USA
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Amanda W Lund
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Keith Syson Chan
- Department of Urology, Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, USA
| | - Jlenia Guarnerio
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- David Geffen Medical School, Department of Medicine, University of California, Los Angeles, CA, USA.
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
83
|
Wang F, Li Z, Xu T, Zhang Q, Ma T, Li S, Wang X. A comprehensive multi-omics analysis identifies a robust scoring system for cancer-associated fibroblasts and intervention targets in colorectal cancer. J Cancer Res Clin Oncol 2024; 150:124. [PMID: 38478111 PMCID: PMC10937804 DOI: 10.1007/s00432-023-05548-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 11/15/2023] [Indexed: 03/17/2024]
Abstract
BACKGROUND Cancer-associated fibroblasts (CAF) play a critical role in promoting tumor growth, metastasis, and immune evasion. While numerous studies have investigated CAF, there remains a paucity of research on their clinical application in colorectal cancer (CRC). METHODS In this study, we collected differentially expressed genes between CAF and normal fibroblasts (NF) from previous CRC studies, and utilized machine learning analysis to differentiate two distinct subtypes of CAF in CRC. To enable practical application, a CAF-related genes (CAFGs) scoring system was developed based on multivariate Cox regression. We then conducted functional enrichment analysis, Kaplan-Meier plot, consensus molecular subtypes (CMS) classification, and Tumor Immune Dysfunction and Exclusion (TIDE) algorithm to investigate the relationship between the CAFGs scoring system and various biological mechanisms, prognostic value, tumor microenvironment, and response to immune checkpoint blockade (ICB) therapy. Moreover, single-cell transcriptomics and proteomics analyses have been employed to validate the significance of scoring system-related molecules in the identity and function of CAF. RESULTS We unveiled significant distinctions in tumor immune status and prognosis not only between the CAF clusters, but also across high and low CAFGs groups. Specifically, patients in CAF cluster 2 or with high CAFGs scores exhibited higher CAF markers and were enriched for CAF-related biological pathways such as epithelial-mesenchymal transition (EMT) and angiogenesis. In addition, CAFGs score was identified as a risk index and correlated with poor overall survival (OS), progression-free survival (PFS), disease-free survival (DFS), and recurrence-free survival (RFS). High CAFGs scores were observed in patients with advanced stages, CMS4, as well as lymphatic invasion. Furthermore, elevated CAFG scores in patients signified a suppressive tumor microenvironment characterized by the upregulation of programmed death-ligand 1 (PD-L1), T-cell dysfunction, exclusion, and TIDE score. And high CAFGs scores can differentiate patients with lower response rates and poor prognosis under ICB therapy. Notably, single-cell transcriptomics and proteomics analyses identified several molecules related to CAF identity and function, such as FSTL1, IGFBP7, and FBN1. CONCLUSION We constructed a robust CAFGs score system with clinical significance using multiple CRC cohorts. In addition, we identified several molecules related to CAF identity and function that could be potential intervention targets for CRC patients.
Collapse
Affiliation(s)
- Feng Wang
- Department of Gastrointestinal Surgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China.
| | - Zhenlin Li
- Department of Surgical Clinical, School of Heze Medical College, Heze, China
| | - Tianlei Xu
- Department of Gastrointestinal Surgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Qian Zhang
- Department of Gastrointestinal Surgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Tianyi Ma
- Department of Gastrointestinal Surgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Sijia Li
- Department of Gastrointestinal Surgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Xiaohui Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
84
|
Zhi Y, Wang Q, Zi M, Zhang S, Ge J, Liu K, Lu L, Fan C, Yan Q, Shi L, Chen P, Fan S, Liao Q, Guo C, Wang F, Gong Z, Xiong W, Zeng Z. Spatial Transcriptomic and Metabolomic Landscapes of Oral Submucous Fibrosis-Derived Oral Squamous Cell Carcinoma and its Tumor Microenvironment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306515. [PMID: 38229179 PMCID: PMC10966560 DOI: 10.1002/advs.202306515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 12/19/2023] [Indexed: 01/18/2024]
Abstract
In South and Southeast Asia, the habit of chewing betel nuts is prevalent, which leads to oral submucous fibrosis (OSF). OSF is a well-established precancerous lesion, and a portion of OSF cases eventually progress to oral squamous cell carcinoma (OSCC). However, the specific molecular mechanisms underlying the malignant transformation of OSCC from OSF are poorly understood. In this study, the leading-edge techniques of Spatial Transcriptomics (ST) and Spatial Metabolomics (SM) are integrated to obtain spatial location information of cancer cells, fibroblasts, and immune cells, as well as the transcriptomic and metabolomic landscapes in OSF-derived OSCC tissues. This work reveals for the first time that some OSF-derived OSCC cells undergo partial epithelial-mesenchymal transition (pEMT) within the in situ carcinoma (ISC) region, eventually acquiring fibroblast-like phenotypes and participating in collagen deposition. Complex interactions among epithelial cells, fibroblasts, and immune cells in the tumor microenvironment are demonstrated. Most importantly, significant metabolic reprogramming in OSF-derived OSCC, including abnormal polyamine metabolism, potentially playing a pivotal role in promoting tumorigenesis and immune evasion is discovered. The ST and SM data in this study shed new light on deciphering the mechanisms of OSF-derived OSCC. The work also offers invaluable clues for the prevention and treatment of OSCC.
Collapse
Affiliation(s)
- Yuan Zhi
- Department of Oral and Maxillofacial SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaHunan410011China
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer MetabolismHunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaHunan410078China
| | - Qian Wang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer MetabolismHunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaHunan410078China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of EducationCancer Research Institute and School of Basic Medicine SciencesCentral South UniversityChangshaHunan410078China
| | - Moxin Zi
- Department of Oral and Maxillofacial SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaHunan410011China
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer MetabolismHunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaHunan410078China
| | - Shanshan Zhang
- Department of StomatologyXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Junshang Ge
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of EducationCancer Research Institute and School of Basic Medicine SciencesCentral South UniversityChangshaHunan410078China
| | - Keyue Liu
- Department of Oral and Maxillofacial SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaHunan410011China
| | - Linsong Lu
- Department of Oral and Maxillofacial SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaHunan410011China
| | - Chunmei Fan
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of EducationCancer Research Institute and School of Basic Medicine SciencesCentral South UniversityChangshaHunan410078China
| | - Qijia Yan
- Department of StomatologyXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Lei Shi
- Department of Oral and Maxillofacial SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaHunan410011China
| | - Pan Chen
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer MetabolismHunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaHunan410078China
| | - Songqing Fan
- Department of Oral and Maxillofacial SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaHunan410011China
| | - Qianjin Liao
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer MetabolismHunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaHunan410078China
| | - Can Guo
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of EducationCancer Research Institute and School of Basic Medicine SciencesCentral South UniversityChangshaHunan410078China
| | - Fuyan Wang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of EducationCancer Research Institute and School of Basic Medicine SciencesCentral South UniversityChangshaHunan410078China
| | - Zhaojian Gong
- Department of Oral and Maxillofacial SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaHunan410011China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of EducationCancer Research Institute and School of Basic Medicine SciencesCentral South UniversityChangshaHunan410078China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer MetabolismHunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaHunan410078China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of EducationCancer Research Institute and School of Basic Medicine SciencesCentral South UniversityChangshaHunan410078China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer MetabolismHunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaHunan410078China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of EducationCancer Research Institute and School of Basic Medicine SciencesCentral South UniversityChangshaHunan410078China
| |
Collapse
|
85
|
Szczepanski JM, Rudolf MA, Shi J. Clinical Evaluation of the Pancreatic Cancer Microenvironment: Opportunities and Challenges. Cancers (Basel) 2024; 16:794. [PMID: 38398185 PMCID: PMC10887250 DOI: 10.3390/cancers16040794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/10/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
Advances in our understanding of pancreatic ductal adenocarcinoma (PDAC) and its tumor microenvironment (TME) have the potential to transform treatment for the hundreds of thousands of patients who are diagnosed each year. Whereas the clinical assessment of cancer cell genetics has grown increasingly sophisticated and personalized, current protocols to evaluate the TME have lagged, despite evidence that the TME can be heterogeneous within and between patients. Here, we outline current protocols for PDAC diagnosis and management, review novel biomarkers, and highlight potential opportunities and challenges when evaluating the PDAC TME as we prepare to translate emerging TME-directed therapies to the clinic.
Collapse
Affiliation(s)
| | | | - Jiaqi Shi
- Department of Pathology and Clinical Labs, University of Michigan, Ann Arbor, MI 48109, USA; (J.M.S.); (M.A.R.)
| |
Collapse
|
86
|
Francescone R, Crawford HC, Vendramini-Costa DB. Rethinking the Roles of Cancer-Associated Fibroblasts in Pancreatic Cancer. Cell Mol Gastroenterol Hepatol 2024; 17:737-743. [PMID: 38316215 PMCID: PMC10966284 DOI: 10.1016/j.jcmgh.2024.01.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/07/2024]
Abstract
Bearing a dismal 5-year survival rate, pancreatic ductal adenocarcinoma (PDAC) is a challenging disease that features a unique fibroinflammatory tumor microenvironment. As major components of the PDAC tumor microenvironment, cancer-associated fibroblasts are still poorly understood and their contribution to the several hallmarks of PDAC, such as resistance to therapies, immunosuppression, and high incidence of metastasis, is likely underestimated. There have been encouraging advances in the understanding of these fascinating cells, but many controversies remain, leaving the field still actively exploring the full scope of their contributions in PDAC progression. Here we pose several important considerations regarding PDAC cancer-associated fibroblast functions. We posit that transcriptomic analyses be interpreted with caution, when aiming to uncover the functional contributions of these cells. Moreover, we propose that normalizing these functions, rather than eliminating them, will provide the opportunity to enhance therapeutic response. Finally, we propose that cancer-associated fibroblasts should not be studied in isolation, but in conjunction with its extracellular matrix, because their respective functions are coordinated and concordant.
Collapse
Affiliation(s)
- Ralph Francescone
- Department of Surgery, Henry Ford Health, Detroit, Michigan; Henry Ford Pancreatic Cancer Center, Henry Ford Health, Detroit, Michigan
| | - Howard C Crawford
- Department of Surgery, Henry Ford Health, Detroit, Michigan; Henry Ford Pancreatic Cancer Center, Henry Ford Health, Detroit, Michigan
| | - Debora Barbosa Vendramini-Costa
- Department of Surgery, Henry Ford Health, Detroit, Michigan; Henry Ford Pancreatic Cancer Center, Henry Ford Health, Detroit, Michigan.
| |
Collapse
|
87
|
Cohen C, Mhaidly R, Croizer H, Kieffer Y, Leclere R, Vincent-Salomon A, Robley C, Anglicheau D, Rabant M, Sannier A, Timsit MO, Eddy S, Kretzler M, Ju W, Mechta-Grigoriou F. WNT-dependent interaction between inflammatory fibroblasts and FOLR2+ macrophages promotes fibrosis in chronic kidney disease. Nat Commun 2024; 15:743. [PMID: 38272907 PMCID: PMC10810789 DOI: 10.1038/s41467-024-44886-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 01/08/2024] [Indexed: 01/27/2024] Open
Abstract
Chronic kidney disease (CKD) is a public health problem driven by myofibroblast accumulation, leading to interstitial fibrosis. Heterogeneity is a recently recognized characteristic in kidney fibroblasts in CKD, but the role of different populations is still unclear. Here, we characterize a proinflammatory fibroblast population (named CXCL-iFibro), which corresponds to an early state of myofibroblast differentiation in CKD. We demonstrate that CXCL-iFibro co-localize with macrophages in the kidney and participate in their attraction, accumulation, and switch into FOLR2+ macrophages from early CKD stages on. In vitro, macrophages promote the switch of CXCL-iFibro into ECM-secreting myofibroblasts through a WNT/β-catenin-dependent pathway, thereby suggesting a reciprocal crosstalk between these populations of fibroblasts and macrophages. Finally, the detection of CXCL-iFibro at early stages of CKD is predictive of poor patient prognosis, which shows that the CXCL-iFibro population is an early player in CKD progression and demonstrates the clinical relevance of our findings.
Collapse
Affiliation(s)
- Camille Cohen
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, rue d'Ulm, Paris, F-75005, France
| | - Rana Mhaidly
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, rue d'Ulm, Paris, F-75005, France
| | - Hugo Croizer
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, rue d'Ulm, Paris, F-75005, France
| | - Yann Kieffer
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, rue d'Ulm, Paris, F-75005, France
| | - Renaud Leclere
- Department of Diagnostic and Theragnostic Medicine, Institut Curie Hospital Group, 26, rue d'Ulm, F-75248, Paris, France
| | - Anne Vincent-Salomon
- Department of Diagnostic and Theragnostic Medicine, Institut Curie Hospital Group, 26, rue d'Ulm, F-75248, Paris, France
| | - Catherine Robley
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, rue d'Ulm, Paris, F-75005, France
| | - Dany Anglicheau
- Department of Nephrology and Kidney Transplantation, Necker Hospital, AP-HP, Paris Cité University, Inserm U1151, 149 rue de Sèvres, 75015, Paris, France
| | - Marion Rabant
- Department of Pathology, Necker Hospital, AP-HP, Paris Cité University, 149 rue de Sèvres, 75015, Paris, France
| | - Aurélie Sannier
- Department of Pathology, AP-HP, Bichat-Claude Bernard Hospital, Paris Cité University, Inserm, U1148, 46, rue Henri Huchard, 75877, Paris, France
| | - Marc-Olivier Timsit
- Department of Urology, Européen George Pompidou Hospital, APHP, Paris Cité University, Paris, France
| | - Sean Eddy
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Matthias Kretzler
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Wenjun Ju
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Fatima Mechta-Grigoriou
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75248, Paris, France.
- Inserm, U830, 26, rue d'Ulm, Paris, F-75005, France.
| |
Collapse
|
88
|
Arebro J, Lee CM, Bennewith KL, Garnis C. Cancer-Associated Fibroblast Heterogeneity in Malignancy with Focus on Oral Squamous Cell Carcinoma. Int J Mol Sci 2024; 25:1300. [PMID: 38279300 PMCID: PMC10816981 DOI: 10.3390/ijms25021300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 01/28/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) remains an understudied and significant global cancer killer and dismal survival rates have not changed in decades. A better understanding of the molecular basis of OSCC progression and metastasis is needed to develop new approaches for treating this disease. The supportive network surrounding cancer tumor cells known as the tumor microenvironment (TME) has gained increasing interest lately since it performs essential protumorigenic functions. Cancer-associated fibroblasts (CAFs) are one of the main cell types in the TME and are known to play a key role in influencing the biological behavior of tumors. CAFs present a heterogeneity both in phenotype as well as functions, leading to the suggestion of different CAF subtypes in several cancer forms. The task to subtype CAFs in OSCC has, however, just begun, and there is today no united way of subtyping CAFs in this disease. This review aims to define the features of CAFs and to summarize CAF subtype research in malignancy with focus on OSCC including aspects as disease prognosis and therapeutic opportunities.
Collapse
Affiliation(s)
- Julia Arebro
- Department of Interdisciplinary Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada; (C.-M.L.); (K.L.B.); (C.G.)
- Division of ENT Diseases, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 14186 Stockholm, Sweden
- Department of ENT Diseases, Karolinska University Hospital, 14186 Stockholm, Sweden
| | - Che-Min Lee
- Department of Interdisciplinary Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada; (C.-M.L.); (K.L.B.); (C.G.)
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V5Z 1L3, Canada
| | - Kevin L. Bennewith
- Department of Interdisciplinary Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada; (C.-M.L.); (K.L.B.); (C.G.)
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V5Z 1L3, Canada
- Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z7, Canada
| | - Cathie Garnis
- Department of Interdisciplinary Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada; (C.-M.L.); (K.L.B.); (C.G.)
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V5Z 1L3, Canada
- Division of Otolaryngology, Department of Surgery, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| |
Collapse
|
89
|
Han H, Qian C, Song M, Zhong C, Zhao Y, Lu Y. Fibroblasts: invigorated targets in pre-metastatic niche formation. Int J Biol Sci 2024; 20:1110-1124. [PMID: 38322116 PMCID: PMC10845297 DOI: 10.7150/ijbs.87680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 01/11/2024] [Indexed: 02/08/2024] Open
Abstract
At present, tumor metastasis still remains the leading contributor to high recurrence and mortality in cancer patients. There have been no clinically effective therapeutic strategies for treating patients with metastatic cancer. In recent years, a growing body of evidence has shown that the pre-metastatic niche (PMN) plays a crucial role in driving tumor metastasis. Nevertheless, a clear and detailed understanding of the formation of PMN is still lacking given the fact that PMN formation involves in a wealth of complicated communications and underlying mechanisms between primary tumors and metastatic target organs. Despite that the roles of numerous components including tumor exosomes and extracellular vesicles in influencing the evolution of PMN have been well documented, the involvement of cancer-associated fibroblasts (CAFs) in the tumor microenvironment for controlling PMN formation is frequently overlooked. It has been increasingly recognized that fibroblasts trigger the formation of PMN by virtue of modulating exosomes, metabolism and so on. In this review, we mainly summarize the underlying mechanisms of fibroblasts from diverse origins in exerting impacts on PMN evolution, and further highlight the prospective strategies for targeting fibroblasts to prevent PMN formation.
Collapse
Affiliation(s)
- Hongkuan Han
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Cheng Qian
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Mengyao Song
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chongjin Zhong
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yang Zhao
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
90
|
Li C, Guo H, Zhai P, Yan M, Liu C, Wang X, Shi C, Li J, Tong T, Zhang Z, Ma H, Zhang J. Spatial and Single-Cell Transcriptomics Reveal a Cancer-Associated Fibroblast Subset in HNSCC That Restricts Infiltration and Antitumor Activity of CD8+ T Cells. Cancer Res 2024; 84:258-275. [PMID: 37930937 PMCID: PMC10790129 DOI: 10.1158/0008-5472.can-23-1448] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 09/14/2023] [Accepted: 11/01/2023] [Indexed: 11/08/2023]
Abstract
Although immunotherapy can prolong survival in some patients with head and neck squamous cell carcinoma (HNSCC), the response rate remains low. Clarification of the critical mechanisms regulating CD8+ T-cell infiltration and dysfunction in the tumor microenvironment could help maximize the benefit of immunotherapy for treating HNSCC. Here, we performed spatial transcriptomic analysis of HNSCC specimens with differing immune infiltration and single-cell RNA sequencing of five pairs of tumor and adjacent tissues, revealing specific cancer-associated fibroblast (CAF) subsets related to CD8+ T-cell infiltration restriction and dysfunction. These CAFs exhibited high expression of CXCLs (CXCL9, CXCL10, and CXCL12) and MHC-I and enrichment of galectin-9 (Gal9). The proportion of MHC-IhiGal9+ CAFs was inversely correlated with abundance of a TCF1+GZMK+ subset of CD8+ T cells. Gal9 on CAFs induced CD8+ T-cell dysfunction and decreased the proportion of tumor-infiltrating TCF1+CD8+ T cells. Collectively, the identification of MHC-IhiGal9+ CAFs advances the understanding of the precise role of CAFs in cancer immune evasion and paves the way for more effective immunotherapy for HNSCC. SIGNIFICANCE Spatial analysis identifies IFN-induced MHC-IhiGal9+ CAFs that form a trap for CD8+ T cells, providing insights into the complex networks in the tumor microenvironment that regulate T-cell infiltration and function.
Collapse
Affiliation(s)
- Chuwen Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- National Center for Stomatology, Shanghai, People's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China
- Shanghai Research Institute of Stomatology, Shanghai, People's Republic of China
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai, People's Republic of China
| | - Haiyan Guo
- Department of Clinical Laboratory, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Peisong Zhai
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- National Center for Stomatology, Shanghai, People's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China
- Shanghai Research Institute of Stomatology, Shanghai, People's Republic of China
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai, People's Republic of China
| | - Ming Yan
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- National Center for Stomatology, Shanghai, People's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China
- Shanghai Research Institute of Stomatology, Shanghai, People's Republic of China
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai, People's Republic of China
| | - Chun Liu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- National Center for Stomatology, Shanghai, People's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China
- Shanghai Research Institute of Stomatology, Shanghai, People's Republic of China
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai, People's Republic of China
| | - Xiaoning Wang
- Department of Oral Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Chaoji Shi
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- National Center for Stomatology, Shanghai, People's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China
- Shanghai Research Institute of Stomatology, Shanghai, People's Republic of China
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai, People's Republic of China
| | - Jiang Li
- Department of Oral Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Tong Tong
- Department of Oral and Maxillofacial Surgery, Shanghai Stomatological Hospital, Fudan University, Shanghai, People's Republic of China
| | - Zhiyuan Zhang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- National Center for Stomatology, Shanghai, People's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China
- Shanghai Research Institute of Stomatology, Shanghai, People's Republic of China
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai, People's Republic of China
- Shanghai Research Institute of Stomatology, Shanghai, People's Republic of China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, People's Republic of China
| | - Hailong Ma
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- National Center for Stomatology, Shanghai, People's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China
- Shanghai Research Institute of Stomatology, Shanghai, People's Republic of China
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai, People's Republic of China
| | - Jianjun Zhang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- National Center for Stomatology, Shanghai, People's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China
- Shanghai Research Institute of Stomatology, Shanghai, People's Republic of China
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai, People's Republic of China
| |
Collapse
|
91
|
Mucciolo G, Araos Henríquez J, Jihad M, Pinto Teles S, Manansala JS, Li W, Ashworth S, Lloyd EG, Cheng PSW, Luo W, Anand A, Sawle A, Piskorz A, Biffi G. EGFR-activated myofibroblasts promote metastasis of pancreatic cancer. Cancer Cell 2024; 42:101-118.e11. [PMID: 38157863 DOI: 10.1016/j.ccell.2023.12.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 10/10/2023] [Accepted: 12/04/2023] [Indexed: 01/03/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis. Cancer-associated fibroblasts (CAFs) are recognized potential therapeutic targets, but poor understanding of these heterogeneous cell populations has limited the development of effective treatment strategies. We previously identified transforming growth factor beta (TGF-β) as a main driver of myofibroblastic CAFs (myCAFs). Here, we show that epidermal growth factor receptor/Erb-B2 receptor (EGFR/ERBB2) signaling is induced by TGF-β in myCAFs through an autocrine process mediated by amphiregulin. Inhibition of this EGFR/ERBB2-signaling network in PDAC organoid-derived cultures and mouse models differentially impacts distinct CAF subtypes, providing insights into mechanisms underpinning their heterogeneity. Remarkably, EGFR-activated myCAFs promote PDAC metastasis in mice, unmasking functional significance in myCAF heterogeneity. Finally, analyses of other cancer datasets suggest that these processes might operate in other malignancies. These data provide functional relevance to myCAF heterogeneity and identify a candidate target for preventing tumor invasion in PDAC.
Collapse
Affiliation(s)
- Gianluca Mucciolo
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK
| | - Joaquín Araos Henríquez
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK
| | - Muntadher Jihad
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK
| | - Sara Pinto Teles
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK
| | - Judhell S Manansala
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK
| | - Wenlong Li
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK
| | - Sally Ashworth
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK
| | - Eloise G Lloyd
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK
| | - Priscilla S W Cheng
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK
| | - Weike Luo
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK
| | - Akanksha Anand
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK
| | - Ashley Sawle
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK
| | - Anna Piskorz
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK
| | - Giulia Biffi
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, Cambridge CB2 0RE, UK.
| |
Collapse
|
92
|
Yan W, Menjivar RE, Bonilla ME, Steele NG, Kemp SB, Du W, Donahue KL, Brown K, Carpenter ES, Avritt FR, Irizarry-Negron VM, Yang S, Burns WR, Zhang Y, di Magliano MP, Bednar F. Notch Signaling Regulates Immunosuppressive Tumor-Associated Macrophage Function in Pancreatic Cancer. Cancer Immunol Res 2024; 12:91-106. [PMID: 37931247 PMCID: PMC10842043 DOI: 10.1158/2326-6066.cir-23-0037] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 07/08/2023] [Accepted: 10/31/2023] [Indexed: 11/08/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDA) continues to have a dismal prognosis. The poor survival of patients with PDA has been attributed to a high rate of early metastasis and low efficacy of current therapies, which partly result from its complex immunosuppressive tumor microenvironment. Previous studies from our group and others have shown that tumor-associated macrophages (TAM) are instrumental in maintaining immunosuppression in PDA. Here, we explored the role of Notch signaling, a key regulator of immune response, within the PDA microenvironment. We identified Notch pathway components in multiple immune cell types within human and mouse pancreatic cancer. TAMs, the most abundant immune cell population in the tumor microenvironment, expressed high levels of Notch receptors, with cognate ligands such as JAG1 expressed on tumor epithelial cells, endothelial cells, and fibroblasts. TAMs with activated Notch signaling expressed higher levels of immunosuppressive mediators, suggesting that Notch signaling plays a role in macrophage polarization within the PDA microenvironment. Genetic inhibition of Notch in myeloid cells led to reduced tumor size and decreased macrophage infiltration in an orthotopic PDA model. Combination of pharmacologic Notch inhibition with PD-1 blockade resulted in increased cytotoxic T-cell infiltration, tumor cell apoptosis, and smaller tumor size. Our work implicates macrophage Notch signaling in the establishment of immunosuppression and indicates that targeting the Notch pathway may improve the efficacy of immune-based therapies in patients with PDA.
Collapse
Affiliation(s)
- Wei Yan
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Rosa E. Menjivar
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Monica E. Bonilla
- Cancer Biology Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nina G. Steele
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Samantha B. Kemp
- Molecular and Cellular Pathology Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Wenting Du
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Katelyn L. Donahue
- Cancer Biology Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kristee Brown
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Eileen S. Carpenter
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor MI 48109, USA
| | - Faith R. Avritt
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Sion Yang
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - William R. Burns
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yaqing Zhang
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Marina Pasca di Magliano
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Filip Bednar
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
- Cancer Biology Program, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
93
|
Luo W, Wen T, Qu X. Tumor immune microenvironment-based therapies in pancreatic ductal adenocarcinoma: time to update the concept. J Exp Clin Cancer Res 2024; 43:8. [PMID: 38167055 PMCID: PMC10759657 DOI: 10.1186/s13046-023-02935-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal solid tumors. The tumor immune microenvironment (TIME) formed by interactions among cancer cells, immune cells, cancer-associated fibroblasts (CAF), and extracellular matrix (ECM) components drives PDAC in a more immunosuppressive direction: this is a major cause of therapy resistance and poor prognosis. In recent years, research has advanced our understanding of the signaling mechanism by which TIME components interact with the tumor and the evolution of immunophenotyping. Through revolutionary technologies such as single-cell sequencing, we have gone from simply classifying PDACs as "cold" and "hot" to a more comprehensive approach of immunophenotyping that considers all the cells and matrix components. This is key to improving the clinical efficacy of PDAC treatments. In this review, we elaborate on various TIME components in PDAC, the signaling mechanisms underlying their interactions, and the latest research into PDAC immunophenotyping. A deep understanding of these network interactions will contribute to the effective combination of TIME-based therapeutic approaches, such as immune checkpoint inhibitors (ICI), adoptive cell therapy, therapies targeting myeloid cells, CAF reprogramming, and stromal normalization. By selecting the appropriate integrated therapies based on precise immunophenotyping, significant advances in the future treatment of PDAC are possible.
Collapse
Affiliation(s)
- Wenyu Luo
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, 110001, Liaoning, China
| | - Ti Wen
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, 110001, China.
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, 110001, Liaoning, China.
| | - Xiujuan Qu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, 110001, China.
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
94
|
Li Z, Pai R, Gupta S, Currenti J, Guo W, Di Bartolomeo A, Feng H, Zhang Z, Li Z, Liu L, Singh A, Bai Y, Yang B, Mishra A, Yang K, Qiao L, Wallace M, Yin Y, Xia Q, Chan JKY, George J, Chow PKH, Ginhoux F, Sharma A. Presence of onco-fetal neighborhoods in hepatocellular carcinoma is associated with relapse and response to immunotherapy. NATURE CANCER 2024; 5:167-186. [PMID: 38168935 DOI: 10.1038/s43018-023-00672-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 10/16/2023] [Indexed: 01/05/2024]
Abstract
Onco-fetal reprogramming of the tumor ecosystem induces fetal developmental signatures in the tumor microenvironment, leading to immunosuppressive features. Here, we employed single-cell RNA sequencing, spatial transcriptomics and bulk RNA sequencing to delineate specific cell subsets involved in hepatocellular carcinoma (HCC) relapse and response to immunotherapy. We identified POSTN+ extracellular matrix cancer-associated fibroblasts (EM CAFs) as a prominent onco-fetal interacting hub, promoting tumor progression. Cell-cell communication and spatial transcriptomics analysis revealed crosstalk and co-localization of onco-fetal cells, including POSTN+ CAFs, FOLR2+ macrophages and PLVAP+ endothelial cells. Further analyses suggest an association between onco-fetal reprogramming and epithelial-mesenchymal transition (EMT), tumor cell proliferation and recruitment of Treg cells, ultimately influencing early relapse and response to immunotherapy. In summary, our study identifies POSTN+ CAFs as part of the HCC onco-fetal niche and highlights its potential influence in EMT, relapse and immunotherapy response, paving the way for the use of onco-fetal signatures for therapeutic stratification.
Collapse
Affiliation(s)
- Ziyi Li
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rhea Pai
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, Perth, Western Australia, Australia
- Curtin Medical School, Curtin University, Perth, Western Australia, Australia
| | - Saurabh Gupta
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, Perth, Western Australia, Australia
- Curtin Medical School, Curtin University, Perth, Western Australia, Australia
| | - Jennifer Currenti
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, Perth, Western Australia, Australia
- Curtin Medical School, Curtin University, Perth, Western Australia, Australia
| | - Wei Guo
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Anna Di Bartolomeo
- Storr Liver Centre, The Westmead Institute for Medical Research and Westmead Hospital, University of Sydney, Sydney, New South Wales, Australia
| | - Hao Feng
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Institute of Transplantation, Shanghai, China
| | - Zijie Zhang
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhizhen Li
- Department of Biliary Tract Surgery I, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Longqi Liu
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, P. R. China
| | - Abhishek Singh
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, Perth, Western Australia, Australia
| | - Yinqi Bai
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, P. R. China
| | | | - Archita Mishra
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
- Telethon Kids Institute, University of Western Australia, Perth Children's Hospital, Nedlands, Western Australia, Australia
| | - Katharine Yang
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Liang Qiao
- Storr Liver Centre, The Westmead Institute for Medical Research and Westmead Hospital, University of Sydney, Sydney, New South Wales, Australia
| | - Michael Wallace
- Department of Hepatology and Western Australian Liver Transplant Service, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
- Medical School, University of Western Australia, Nedlands, Western Australia, Australia
| | - Yujia Yin
- Department of Obstetrics and Gynecology, Xinhua Hospital Affiliated to Shanghai Jiaotong University Medicine School, Shanghai, China
| | - Qiang Xia
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Institute of Transplantation, Shanghai, China
| | - Jerry Kok Yen Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore
- Academic Clinical Program in Obstetrics and Gynaecology, Duke-NUS Medical School, Singapore, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jacob George
- Storr Liver Centre, The Westmead Institute for Medical Research and Westmead Hospital, University of Sydney, Sydney, New South Wales, Australia
| | - Pierce Kah-Hoe Chow
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital and National Cancer Centre Singapore, Singapore, Singapore.
- Surgery Academic Clinical Programme, Duke-NUS Medical School, Singapore, Singapore.
| | - Florent Ginhoux
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore.
- Gustave Roussy Cancer Campus, Villejuif, France.
| | - Ankur Sharma
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, Perth, Western Australia, Australia.
- Curtin Medical School, Curtin University, Perth, Western Australia, Australia.
- Institute of Molecular and Cell Biology, A∗STAR, Singapore, Singapore.
- KK Research Centre, KK Women's and Children's Hospital, Singapore, Singapore.
| |
Collapse
|
95
|
Gu Y, Chen Q, Yin H, Zeng M, Gao S, Wang X. Cancer-associated fibroblasts in neoadjuvant setting for solid cancers. Crit Rev Oncol Hematol 2024; 193:104226. [PMID: 38056580 DOI: 10.1016/j.critrevonc.2023.104226] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/15/2023] [Accepted: 11/30/2023] [Indexed: 12/08/2023] Open
Abstract
Therapeutic approaches for cancer have become increasingly diverse in recent times. A comprehensive understanding of the tumor microenvironment (TME) holds great potential for enhancing the precision of tumor therapies. Neoadjuvant therapy offers the possibility of alleviating patient symptoms and improving overall quality of life. Additionally, it may facilitate the reduction of inoperable tumors and prevent potential preoperative micrometastases. Within the TME, cancer-associated fibroblasts (CAFs) play a prominent role as they generate various elements that contribute to tumor progression. Particularly, extracellular matrix (ECM) produced by CAFs prevents immune cell infiltration into the TME, hampers drug penetration, and diminishes therapeutic efficacy. Therefore, this review provides a summary of the heterogeneity and interactions of CAFs within the TME, with a specific focus on the influence of neoadjuvant therapy on the microenvironment, particularly CAFs. Finally, we propose several potential and promising therapeutic strategies targeting CAFs, which may efficiently eliminate CAFs to decrease stroma density and impair their functions.
Collapse
Affiliation(s)
- Yanan Gu
- Department of Radiology, Zhongshan Hospital and Shanghai Institute of Medical Imaging, Fudan University, Shanghai 200032, China; Department of Interventional Radiology, Zhongshan Hospital Fudan University Shanghai, 200032, China
| | - Qiangda Chen
- Department of Pancreatic Surgery, Zhongshan Hospital Fudan University, Shanghai 200032, China; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hanlin Yin
- Department of Pancreatic Surgery, Zhongshan Hospital Fudan University, Shanghai 200032, China; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Mengsu Zeng
- Department of Radiology, Zhongshan Hospital and Shanghai Institute of Medical Imaging, Fudan University, Shanghai 200032, China
| | - Shanshan Gao
- Department of Radiology, Zhongshan Hospital and Shanghai Institute of Medical Imaging, Fudan University, Shanghai 200032, China.
| | - Xiaolin Wang
- Department of Radiology, Zhongshan Hospital and Shanghai Institute of Medical Imaging, Fudan University, Shanghai 200032, China; Department of Interventional Radiology, Zhongshan Hospital Fudan University Shanghai, 200032, China.
| |
Collapse
|
96
|
Pei S, Sjölund J, Pan Y, Pietras K, Karlsson MCI. Cancer-associated fibroblasts express CD1d and activate invariant natural killer T cells under cellular stress. Cell Mol Immunol 2024; 21:91-94. [PMID: 37735500 PMCID: PMC10757709 DOI: 10.1038/s41423-023-01082-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/30/2023] [Indexed: 09/23/2023] Open
Affiliation(s)
- Shengduo Pei
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jonas Sjölund
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University Cancer Centre, Lund University, Lund, Sweden
| | - Yueyun Pan
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Kristian Pietras
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University Cancer Centre, Lund University, Lund, Sweden
| | - Mikael C I Karlsson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
97
|
Affiliation(s)
| | - Claus Jorgensen
- Systems Oncology, Cancer Research UK Manchester Institute, Manchester, UK
| |
Collapse
|
98
|
Liu Z, Zhang Y, Wu C. Single-cell sequencing in pancreatic cancer research: A deeper understanding of heterogeneity and therapy. Biomed Pharmacother 2023; 168:115664. [PMID: 37837881 DOI: 10.1016/j.biopha.2023.115664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/28/2023] [Accepted: 10/06/2023] [Indexed: 10/16/2023] Open
Abstract
Pancreatic cancer, including pancreatic ductal adenocarcinomas (PDACs), is a malignant tumor with characteristics of tumor-stroma interactions. Patients often have a poor prognosis and a poor long-term survival rate. In recent years, rapidly-developing single-cell sequencing techniques have been used to analyze cell populations at a single-cell resolution, so that it is now possible to have a more in-depth and clearer understanding of the genetic composition of pancreatic cancer. In this review, we provide an overview of the current single-cell sequencing techniques and their applications in the exploration of intratumoral heterogeneity, the tumor microenvironment, therapy resistance, and novel treatments. Our hope is to provide new insight into the potential of precision therapy, which will perhaps one day lead to significant advances in PDAC treatment.
Collapse
Affiliation(s)
- Zhuomiao Liu
- Department of Radiation Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Yalin Zhang
- Department of Radiation Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Chunli Wu
- Department of Radiation Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
99
|
Zhou Z, Zhong Y, Zhang Z, Ren X. Spatial transcriptomics deconvolution at single-cell resolution using Redeconve. Nat Commun 2023; 14:7930. [PMID: 38040768 PMCID: PMC10692090 DOI: 10.1038/s41467-023-43600-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/14/2023] [Indexed: 12/03/2023] Open
Abstract
Computational deconvolution with single-cell RNA sequencing data as reference is pivotal to interpreting spatial transcriptomics data, but the current methods are limited to cell-type resolution. Here we present Redeconve, an algorithm to deconvolute spatial transcriptomics data at single-cell resolution, enabling interpretation of spatial transcriptomics data with thousands of nuanced cell states. We benchmark Redeconve with the state-of-the-art algorithms on diverse spatial transcriptomics platforms and datasets and demonstrate the superiority of Redeconve in terms of accuracy, resolution, robustness, and speed. Application to a human pancreatic cancer dataset reveals cancer-clone-specific T cell infiltration, and application to lymph node samples identifies differential cytotoxic T cells between IgA+ and IgG+ spots, providing novel insights into tumor immunology and the regulatory mechanisms underlying antibody class switch.
Collapse
Affiliation(s)
- Zixiang Zhou
- Changping Laboratory, Yard 28, Science Park Road, Changping District, Beijing, China
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, 100871, Beijing, China
| | - Yunshan Zhong
- Changping Laboratory, Yard 28, Science Park Road, Changping District, Beijing, China
| | - Zemin Zhang
- Changping Laboratory, Yard 28, Science Park Road, Changping District, Beijing, China
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, 100871, Beijing, China
| | - Xianwen Ren
- Changping Laboratory, Yard 28, Science Park Road, Changping District, Beijing, China.
| |
Collapse
|
100
|
Shi Y, Guo Y, Zhou J, Cui G, Cheng J, Wu Y, Zhao Y, Fang L, Han X, Yang Y, Sun Y. A spatiotemporal gene expression and cell atlases of the developing rat ovary. Cell Prolif 2023; 56:e13516. [PMID: 37309718 PMCID: PMC10693188 DOI: 10.1111/cpr.13516] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 06/14/2023] Open
Abstract
Normal ovarian development is necessary for the production of healthy oocytes. However, the characteristics of oocytes development at different stages and the regulatory relationship between oocytes and somatic cells remain to be fully explained. Here, we combined scRNA-seq and spatial transcriptomic sequencing to profile the transcriptomic atlas of developing ovarian of the rat. We identified four components from developing granulosa cells including cumulus, primitive, mural, and luteal cells, and constructed their differential transcriptional regulatory networks. Several novel growth signals from oocytes to cumulus cells were identified, such as JAG1-NOTCH2 and FGF9-FGFR2. Moreover, we observed three cumulus sequential phases during follicle development determined by the key transcriptional factors in each cumulus phase (Bckaf1, Gata6, Cebpb, etc.), as well as the potential pinpointed roles of macrophages in luteal regression. Altogether, the single-cell spatial transcriptomic profile of the ovary provides not only a new research dimension for temporal and spatial analysis of ovary development, but also valuable data resources and a research basis for in-depth excavation of the mechanisms of mammalian ovary development.
Collapse
Affiliation(s)
- Yong Shi
- Henan Key Laboratory of Reproduction and Genetics, Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Academy of medical sciencesZhengzhou UniversityZhengzhouChina
| | - Yanjie Guo
- Henan Key Laboratory of Reproduction and Genetics, Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jiayi Zhou
- Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, College of Future Technology, Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- China National Center for BioinformationBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Guanshen Cui
- Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, College of Future Technology, Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- China National Center for BioinformationBeijingChina
| | - Jung‐Chien Cheng
- Henan Key Laboratory of Reproduction and Genetics, Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Ying Wu
- Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, College of Future Technology, Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- China National Center for BioinformationBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yong‐Liang Zhao
- Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, College of Future Technology, Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- China National Center for BioinformationBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Lanlan Fang
- Henan Key Laboratory of Reproduction and Genetics, Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Xiao Han
- Henan Key Laboratory of Reproduction and Genetics, Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, College of Future Technology, Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- China National Center for BioinformationBeijingChina
| | - Yun‐Gui Yang
- Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, College of Future Technology, Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- China National Center for BioinformationBeijingChina
- University of Chinese Academy of SciencesBeijingChina
- Institute of Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
| | - Yingpu Sun
- Henan Key Laboratory of Reproduction and Genetics, Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|